Bio


Uta Francke is professor of Genetics Emerita and Professor of Pediatrics - Medical Genetics at Stanford University. Her research has ranged from human and mouse chromosome identification and gene mapping to the discovery of genes involved in heritable disorders, studies of their functions and of disease-causing mechanisms. Applying genomic technologies to mammalian genetics research, her laboratory developed mouse models for human microdeletions, such as Prader-Willi and Williams-Beuren syndrome.

Dr. Francke holds an M.D. from the University of Munich, Germany. She is board-certified in Pediatrics, and in Clinical and Molecular Genetics and Cytogenetics. She has been an Investigator of the Howard Hughes Medical Institute and the recipient of the Antoine Marfan Award from the National Marfan Foundation, the Colonel Harland Sanders Lifetime Achievement Award in Genetics from the March of Dimes Birth Defects Foundation and the William Allan Award from the American Society of Human Genetics. In 2014,, she received the Association for Molecular Pathology Award for Excellence in Molecular Diagnostics. She has been elected to the Institute of Medicine of the National Academies, the American Association for Advancement of Science and the American Academy of Arts and Sciences. She is a past-president of the American Society for Human Genetics and of the International Federation of Human Genetics Societies, and a founding member of the American College of Medical Genetics.
Dr. Francke teaches medical and molecular genetics and sees patients in the Multidisciplinary Cardiovascular Connective Tissue Disorders Clinic at Stanford.

Dr. Francke has served as a consultant to 23andMe.Inc since 2007, and as a part-time employee from 2010-2013. In her role a Senior Medical Director she was involved in the preparation of health report and in various research projects, foremost the study of consumer responses to receiving BRCA mutation results.

Clinical Focus


  • Neurogenetics
  • Clinical Genetics
  • Marfan syndrome
  • Williams syndrome
  • Heritable connective tissue disorders

Academic Appointments


Honors & Awards


  • Original Member, Highly Cited Researchers database, ISI (2002)
  • Antoine Marfan Award, National Marfan Foundation (1996)
  • President, International Federation of Human Genetics Societies (2000-2002)
  • Elected President, American Society of Human Genetics (1999)
  • Elected Member, American Academy of Arts and Sciences (1997)
  • Elected Fellow, American Association for the Advancement of Science (1995)
  • Elected Member, Institute of Medicine (National Academies) (1990)
  • Elected Associate Member, European Molecular Biology Organization (2009)
  • Colonel Harland Sanders Lifetime Achievement Award in Genetics, March of Dimes Birth Defects Foundation (2001)
  • William Allan Award, American Society for Human Genetics (2012)
  • Award for Excellence in Molecular Diagnostics, Association for Molecular Pathology (2014)

Boards, Advisory Committees, Professional Organizations


  • Chair, Awards Committee, American Society of Human Genetics (2020 - Present)
  • Founding Member, American College of Medical Genetics (1993 - Present)

Professional Education


  • Board Certification: American Board of Medical Genetics and Genomics, Clinical Cytogenetics and Genomics (1982)
  • Board Certification: American Board of Medical Genetics and Genomics, Clinical Genetics (1982)
  • Board Certification: American Board of Pediatrics, Pediatrics (1981)
  • Fellowship: UCSD Medical Genetics Fellowship (1973) CA
  • Fellowship: UCLA Medical Genetics and Genomics Fellowship (1971) CA
  • Residency: Childrens Hospital Los Angeles Pediatric Residency (1970) CA
  • Internship: Klinikum Rechts Der Isar (1969) Germany
  • Medical Education: Ludwig Maximilians Universitat Munchen (1966) Germany
  • Board certification, Clinical Molecular Genetics, American Board of Medical Genetics (2009)
  • M.D. (Dr. med.), Universitaet Munchen, Germany, Medicine (1967)

Current Research and Scholarly Interests


My laboratory at Stanford is closed. My ongoing research activities are collaborative.

2023-24 Courses


Graduate and Fellowship Programs


All Publications


  • Direct-to-consumer genetic testing for factor V Leiden and prothrombin 20210G>A: the consumer experience. Molecular genetics & genomic medicine Elson, S. L., Furlotte, N. A., Hromatka, B. S., Wilson, C. H., Mountain, J. L., Rowbotham, H. M., Varga, E. A., Francke, U. 2020: e1468

    Abstract

    BACKGROUND: Clinical genetic testing for inherited predisposition to venous thromboembolism (VTE) is common among patients and their families. However, there is incomplete consensus about which individuals should receive testing, and the relative risks and benefits.METHODS: We assessed outcomes of receiving direct-to-consumer (DTC) results for the two most common genetic risk factors for VTE, factor V Leiden in the F5 gene (FVL) and prothrombin 20210G>A in the F2 gene (PT). Two thousand three hundred fifty-four customers (1244 variant-positive and 1110 variant-negative individuals) of the personal genetics company 23andMe, Inc., who had received results online for F5 and F2 variants, participated in an online survey-based study. Participants responded to questions about perception of VTE risk, discussion of results with healthcare providers (HCPs) and recommendations received, actions taken to control risk, emotional responses to receiving risk results, and perceived value of the information.RESULTS: Most participants (90% of variant-positive individuals, 99% of variant-negative individuals) had not previously been tested for F5 and/or F2 variants. The majority of variant-positive individuals correctly perceived that they were at higher than average risk for developing VTE. These individuals reported moderate rates of discussing results with HCPs (41%); receiving prevention advice from HCPs (31%), and making behavioral changes to control risk (e.g., exercising more, 30%). A minority (36%) of variant-positive individuals worried more after receiving VTE results. Nevertheless, most participants reported that knowing their risk had been an advantage (78% variant-positive and 58% variant-negative) and were satisfied knowing their genetic probability for VTE (81% variant-positive and 67% variant-negative).CONCLUSION: Consumers reported moderate rates of behavioral change and perceived personal benefit from receiving DTC genetic results for VTE risk.

    View details for DOI 10.1002/mgg3.1468

    View details for PubMedID 32940023

  • Report of the Phenotype of a Patient with Roberts Syndrome and a Rare ESCO2 Variant. Journal of pediatric genetics da Costa Almeida, C. B., Welter, A. T., Abech, G. D., Brandão, G. R., Flores, J. A., Schüle, B. n., Francke, U. n., Fiegenbaum, M. n., Zen, P. R., Rosa, R. F. 2020; 9 (1): 58–62

    Abstract

    Roberts syndrome is a rare autosomal recessive genetic disease. In this report, we report a Brazilian patient with a rare ESCO2 variant. The patient manifested a broad range of clinical findings including the significant, bilateral shortening of the extremities. He deteriorated and passed away at 20 days of age. High-resolution GTG-banded karyotype showed lack of centromeric constriction in some chromosomes, premature centromere separation in others, and repulsion of the heterochromatin regions. Molecular analysis of the ESCO2 gene revealed a deletion of 4 bp involving exon 4 in homozygosity (NM_00107420.2:c.875_878delACAG), which causes loss of ESCO2 function. We describe the clinical presentation caused by a rare ESCO2 variant.

    View details for DOI 10.1055/s-0039-1696636

    View details for PubMedID 31976146

    View details for PubMedCentralID PMC6976314

  • Isolation of a Novel Gene Mutated in Wiskott-Aldrich Syndrome JOURNAL OF IMMUNOLOGY Derry, J. J., Ochs, H. D., Francke, U. 2018; 200 (11): 3671–80

    View details for Web of Science ID 000442385700002

    View details for PubMedID 29784763

  • Germ line variants predispose to both JAK2 V617F clonal hematopoiesis and myeloproliferative neoplasms. Blood Hinds, D. A., Barnholt, K. E., Mesa, R. A., Kiefer, A. K., Do, C. B., Eriksson, N., Mountain, J. L., Francke, U., Tung, J. Y., Nguyen, H. M., Zhang, H., Gojenola, L., Zehnder, J. L., Gotlib, J. 2016; 128 (8): 1121-1128

    Abstract

    We conducted a genome-wide association study (GWAS) to identify novel predisposition alleles associated with Philadelphia chromosome-negative myeloproliferative neoplasms (MPNs) and JAK2 V617F clonal hematopoiesis in the general population. We recruited a web-based cohort of 726 individuals with polycythemia vera, essential thrombocythemia, and myelofibrosis and 252 637 population controls unselected for hematologic phenotypes. Using a single-nucleotide polymorphism (SNP) array platform with custom probes for the JAK2 V617F mutation (V617F), we identified 497 individuals (0.2%) among the population controls who were V617F carriers. We performed a combined GWAS of the MPN cases plus V617F carriers in the control population (n = 1223) vs the remaining controls who were noncarriers for V617F (n = 252 140). For these MPN cases plus V617F carriers, we replicated the germ line JAK2 46/1 haplotype (rs59384377: odds ratio [OR] = 2.4, P = 6.6 × 10(-89)), previously associated with V617F-positive MPN. We also identified genome-wide significant associations in the TERT gene (rs7705526: OR = 1.8, P = 1.1 × 10(-32)), in SH2B3 (rs7310615: OR = 1.4, P = 3.1 × 10(-14)), and upstream of TET2 (rs1548483: OR = 2.0, P = 2.0 × 10(-9)). These associations were confirmed in a separate replication cohort of 446 V617F carriers vs 169 021 noncarriers. In a joint analysis of the combined GWAS and replication results, we identified additional genome-wide significant predisposition alleles associated with CHEK2, ATM, PINT, and GFI1B All SNP ORs were similar for MPN patients and controls who were V617F carriers. These data indicate that the same germ line variants endow individuals with a predisposition not only to MPN, but also to JAK2 V617F clonal hematopoiesis, a more common phenomenon that may foreshadow the development of an overt neoplasm.

    View details for DOI 10.1182/blood-2015-06-652941

    View details for PubMedID 27365426

  • A genome-wide association meta-analysis of self-reported allergy identifies shared and allergy-specific susceptibility loci NATURE GENETICS Hinds, D. A., McMahon, G., Kiefer, A. K., Do, C. B., Eriksson, N., Evans, D. M., St Pourcain, B., Ring, S. M., Mountain, J. L., Francke, U., Davey-Smith, G., Timpson, N. J., Tung, J. Y. 2013; 45 (8): 907-U292

    Abstract

    Allergic disease is very common and carries substantial public-health burdens. We conducted a meta-analysis of genome-wide associations with self-reported cat, dust-mite and pollen allergies in 53,862 individuals. We used generalized estimating equations to model shared and allergy-specific genetic effects. We identified 16 shared susceptibility loci with association P < 5 × 10(-8), including 8 loci previously associated with asthma, as well as 4p14 near TLR1, TLR6 and TLR10 (rs2101521, P = 5.3 × 10(-21)); 6p21.33 near HLA-C and MICA (rs9266772, P = 3.2 × 10(-12)); 5p13.1 near PTGER4 (rs7720838, P = 8.2 × 10(-11)); 2q33.1 in PLCL1 (rs10497813, P = 6.1 × 10(-10)), 3q28 in LPP (rs9860547, P = 1.2 × 10(-9)); 20q13.2 in NFATC2 (rs6021270, P = 6.9 × 10(-9)), 4q27 in ADAD1 (rs17388568, P = 3.9 × 10(-8)); and 14q21.1 near FOXA1 and TTC6 (rs1998359, P = 4.8 × 10(-8)). We identified one locus with substantial evidence of differences in effects across allergies at 6p21.32 in the class II human leukocyte antigen (HLA) region (rs17533090, P = 1.7 × 10(-12)), which was strongly associated with cat allergy. Our study sheds new light on the shared etiology of immune and autoimmune disease.

    View details for DOI 10.1038/ng.2686

    View details for Web of Science ID 000322374900014

    View details for PubMedID 23817569

  • How will genomic information become integrated into the health care system? Molecular genetics & genomic medicine Francke, U. 2013; 1 (2): 67-70

    View details for DOI 10.1002/mgg3.26

    View details for PubMedID 24498603

    View details for PubMedCentralID PMC3865570

  • Genome-Wide Association Analysis Implicates Elastic Microfibrils in the Development of Non-Syndromic Striae Distensae. The Journal of investigative dermatology Tung, J. Y., Kiefer, A. K., Mullins, M., Francke, U., Eriksson, N. 2013

    View details for DOI 10.1038/jid.2013.196

    View details for PubMedID 23633020

  • 2012 William Allan Award: Adventures in Cytogenetics AMERICAN JOURNAL OF HUMAN GENETICS Francke, U. 2013; 92 (3): 325-337

    View details for DOI 10.1016/j.ajhg.2013.01.010

    View details for Web of Science ID 000316161700004

    View details for PubMedID 23472754

    View details for PubMedCentralID PMC3591841

  • Dealing with the unexpected: consumer responses to direct-access BRCA mutation testing PEERJ Francke, U., Dijamco, C., Kiefer, A. K., Eriksson, N., Moiseff, B., Tung, J. Y., Mountain, J. L. 2013; 1

    View details for DOI 10.7717/peerj.8

    View details for Web of Science ID 000209185300007

  • Genome-Wide Analysis Points to Roles for Extracellular Matrix Remodeling, the Visual Cycle, and Neuronal Development in Myopia PLOS GENETICS Kiefer, A. K., Tung, J. Y., Do, C. B., Hinds, D. A., Mountain, J. L., Francke, U., Eriksson, N. 2013; 9 (2)

    Abstract

    Myopia, or nearsightedness, is the most common eye disorder, resulting primarily from excess elongation of the eye. The etiology of myopia, although known to be complex, is poorly understood. Here we report the largest ever genome-wide association study (45,771 participants) on myopia in Europeans. We performed a survival analysis on age of myopia onset and identified 22 significant associations ([Formula: see text]), two of which are replications of earlier associations with refractive error. Ten of the 20 novel associations identified replicate in a separate cohort of 8,323 participants who reported if they had developed myopia before age 10. These 22 associations in total explain 2.9% of the variance in myopia age of onset and point toward a number of different mechanisms behind the development of myopia. One association is in the gene PRSS56, which has previously been linked to abnormally small eyes; one is in a gene that forms part of the extracellular matrix (LAMA2); two are in or near genes involved in the regeneration of 11-cis-retinal (RGR and RDH5); two are near genes known to be involved in the growth and guidance of retinal ganglion cells (ZIC2, SFRP1); and five are in or near genes involved in neuronal signaling or development. These novel findings point toward multiple genetic factors involved in the development of myopia and suggest that complex interactions between extracellular matrix remodeling, neuronal development, and visual signals from the retina may underlie the development of myopia in humans.

    View details for DOI 10.1371/journal.pgen.1003299

    View details for Web of Science ID 000315638300050

    View details for PubMedID 23468642

  • Correlation of Symptom Assessment with Genotyping Analysis of Saliva Samples in a Large Cohort of Myeloproliferative Neoplasm Patients 54th Annual Meeting and Exposition of the American-Society-of-Hematology (ASH) Nguyen, H. (., Hinds, D. A., Barnholt, K. E., Kiefer, A. K., Do, C. B., Eriksson, N., Mountain, J. L., Francke, U., Tung, J. A., Levine, R. L., Zehnder, J. L., Gotlib, J., Mesa, R. A. AMER SOC HEMATOLOGY. 2012
  • Estimation of JAK2 V617F Prevalence by Detection of the Mutation in Saliva Samples From Online MPN and General Population Cohorts 54th Annual Meeting and Exposition of the American-Society-of-Hematology (ASH) Barnholt, K. E., Hinds, D. A., Kiefer, A. K., Do, C. B., Eriksson, N., Mountain, J. L., Francke, U., Tung, J. A., Nguyen, H. (., Levine, R. L., Mesa, R. A., Gotlib, J., Zehnder, J. L. AMER SOC HEMATOLOGY. 2012
  • Comparison of Family History and SNPs for Predicting Risk of Complex Disease PLOS GENETICS Do, C. B., Hinds, D. A., Francke, U., Eriksson, N. 2012; 8 (10)

    Abstract

    The clinical utility of family history and genetic tests is generally well understood for simple Mendelian disorders and rare subforms of complex diseases that are directly attributable to highly penetrant genetic variants. However, little is presently known regarding the performance of these methods in situations where disease susceptibility depends on the cumulative contribution of multiple genetic factors of moderate or low penetrance. Using quantitative genetic theory, we develop a model for studying the predictive ability of family history and single nucleotide polymorphism (SNP)-based methods for assessing risk of polygenic disorders. We show that family history is most useful for highly common, heritable conditions (e.g., coronary artery disease), where it explains roughly 20%-30% of disease heritability, on par with the most successful SNP models based on associations discovered to date. In contrast, we find that for diseases of moderate or low frequency (e.g., Crohn disease) family history accounts for less than 4% of disease heritability, substantially lagging behind SNPs in almost all cases. These results indicate that, for a broad range of diseases, already identified SNP associations may be better predictors of risk than their family history-based counterparts, despite the large fraction of missing heritability that remains to be explained. Our model illustrates the difficulty of using either family history or SNPs for standalone disease prediction. On the other hand, we show that, unlike family history, SNP-based tests can reveal extreme likelihood ratios for a relatively large percentage of individuals, thus providing potentially valuable adjunctive evidence in a differential diagnosis.

    View details for DOI 10.1371/journal.pgen.1002973

    View details for Web of Science ID 000310528400013

    View details for PubMedID 23071447

  • Genetic variants associated with breast size also influence breast cancer risk BMC MEDICAL GENETICS Eriksson, N., Benton, G. M., Do, C. B., Kiefer, A. K., Mountain, J. L., Hinds, D. A., Francke, U., Tung, J. Y. 2012; 13

    Abstract

    While some factors of breast morphology, such as density, are directly implicated in breast cancer, the relationship between breast size and cancer is less clear. Breast size is moderately heritable, yet the genetic variants leading to differences in breast size have not been identified.To investigate the genetic factors underlying breast size, we conducted a genome-wide association study (GWAS) of self-reported bra cup size, controlling for age, genetic ancestry, breast surgeries, pregnancy history and bra band size, in a cohort of 16,175 women of European ancestry.We identified seven single-nucleotide polymorphisms (SNPs) significantly associated with breast size (p<5.10(-8)): rs7816345 near ZNF703, rs4849887 and (independently) rs17625845 flanking INHBB, rs12173570 near ESR1, rs7089814 in ZNF365, rs12371778 near PTHLH, and rs62314947 near AREG. Two of these seven SNPs are in linkage disequilibrium (LD) with SNPs associated with breast cancer (those near ESR1 and PTHLH), and a third (ZNF365) is near, but not in LD with, a breast cancer SNP. The other three loci (ZNF703, INHBB, and AREG) have strong links to breast cancer, estrogen regulation, and breast development.These results provide insight into the genetic factors underlying normal breast development and show that some of these factors are shared with breast cancer. While these results do not directly support any possible epidemiological relationships between breast size and cancer, this study may contribute to a better understanding of the subtle interactions between breast morphology and breast cancer risk.

    View details for DOI 10.1186/1471-2350-13-53

    View details for Web of Science ID 000310354300001

    View details for PubMedID 22747683

  • The new Ghent criteria for Marfan syndrome: what do they change? CLINICAL GENETICS Faivre, L., Collod-Beroud, G., Ades, L., Arbustini, E., Child, A., Callewaert, B. L., Loeys, B., Binquet, C., Gautier, E., Mayer, K., Arslan-Kirchner, M., Grasso, M., Beroud, C., Hamroun, D., Bonithon-Kopp, C., Plauchu, H., Robinson, P. N., De Backer, J., Couckek, P., FRANCKE, U., Bouchot, O., Wolf, J. E., Stheneur, C., Hanna, N., Detaint, D., De Paepe, A., Boileau, C., Jondeauv, G. 2012; 81 (5): 433-442

    Abstract

    The diagnosis of Marfan syndrome (MFS) is challenging and international criteria have been proposed. The 1996 Ghent criteria were adopted worldwide, but new diagnostic criteria for MFS were released in 2010, giving more weight to aortic root aneurysm and ectopia lentis. We aimed to compare the diagnosis reached by applying this new nosology vs the Ghent nosology in a well-known series of 1009 probands defined by the presence of an FBN1 mutation. A total of 842 patients could be classified as MFS according to the new nosology (83%) as compared to 894 (89%) according to the 1996 Ghent criteria. The remaining 17% would be classified as ectopia lentis syndrome (ELS), mitral valve prolapse syndrome or mitral valve, aorta, skeleton and skin (MASS) syndrome, or potential MFS in patients aged less than 20 years. Taking into account the median age at last follow-up (29 years), the possibility has to be considered that these patients would go on to develop classic MFS with time. Although the number of patients for a given diagnosis differed only slightly, the new nosology led to a different diagnosis in 15% of cases. Indeed, 10% of MFS patients were reclassified as ELS or MASS in the absence of aortic dilatation; conversely, 5% were reclassified as MFS in the presence of aortic dilatation. The nosology is easier to apply because the systemic score is helpful to reach the diagnosis of MFS only in a minority of patients. Diagnostic criteria should be a flexible and dynamic tool so that reclassification of patients with alternative diagnosis is possible, requiring regular clinical and aortic follow-up.

    View details for DOI 10.1111/j.1399-0004.2011.01703.x

    View details for Web of Science ID 000302617700007

    View details for PubMedID 21564093

  • Novel Associations for Hypothyroidism Include Known Autoimmune Risk Loci PLOS ONE Eriksson, N., Tung, J. Y., Kiefer, A. K., Hinds, D. A., Francke, U., Mountain, J. L., Do, C. B. 2012; 7 (4)

    Abstract

    Hypothyroidism is the most common thyroid disorder, affecting about 5% of the general population. Here we present the current largest genome-wide association study of hypothyroidism, in 3,736 cases and 35,546 controls. Hypothyroidism was assessed via web-based questionnaires. We identify five genome-wide significant associations, three of which are well known to be involved in a large spectrum of autoimmune diseases: rs6679677 near PTPN22, rs3184504 in SH2B3, and rs2517532 in the HLA class I region (p-values 2.8·10(-13), 2.6·10(-12), and 1.3·10(-8), respectively). We also report associations with rs4915077 near VAV3 (p-value 7.5·10(-10)) and rs925489 near FOXE1 (p value 2.4·10(-19)). VAV3 is involved in immune function, and FOXE1 and PTPN22 have previously been associated with hypothyroidism. Although the HLA class I region and SH2B3 have previously been linked with a number of autoimmune diseases, this is the first report of their association with thyroid disease. The VAV3 association is also novel. We also show suggestive evidence of association for hypothyroidism with a SNP in the HLA class II region (independent of the other HLA association) as well as SNPs in CAPZB, PDE8B, and CTLA4. CAPZB and PDE8B have been linked to TSH levels and CTLA4 to a variety of autoimmune diseases. These results suggest heterogeneity in the genetic etiology of hypothyroidism, implicating genes involved in both autoimmune disorders and thyroid function. Using a genetic risk profile score based on the top association from each of the five genome-wide significant regions in our study, the relative risk between the highest and lowest deciles of genetic risk is 2.0.

    View details for DOI 10.1371/journal.pone.0034442

    View details for Web of Science ID 000305012700033

    View details for PubMedID 22493691

  • Reduction of NADPH-Oxidase Activity Ameliorates the Cardiovascular Phenotype in a Mouse Model of Williams-Beuren Syndrome PLOS GENETICS Campuzano, V., Segura-Puimedon, M., Terrado, V., Sanchez-Rodriguez, C., Coustets, M., Menacho-Marquez, M., Nevado, J., Bustelo, X. R., Francke, U., Perez-Jurado, L. A. 2012; 8 (2)

    Abstract

    A hallmark feature of Williams-Beuren Syndrome (WBS) is a generalized arteriopathy due to elastin deficiency, presenting as stenoses of medium and large arteries and leading to hypertension and other cardiovascular complications. Deletion of a functional NCF1 gene copy has been shown to protect a proportion of WBS patients against hypertension, likely through reduced NADPH-oxidase (NOX)-mediated oxidative stress. DD mice, carrying a 0.67 Mb heterozygous deletion including the Eln gene, presented with a generalized arteriopathy, hypertension, and cardiac hypertrophy, associated with elevated angiotensin II (angII), oxidative stress parameters, and Ncf1 expression. Genetic (by crossing with Ncf1 mutant) and/or pharmacological (with ang II type 1 receptor blocker, losartan, or NOX inhibitor apocynin) reduction of NOX activity controlled hormonal and biochemical parameters in DD mice, resulting in normalized blood pressure and improved cardiovascular histology. We provide strong evidence for implication of the redox system in the pathophysiology of the cardiovascular disease in a mouse model of WBS. The phenotype of these mice can be ameliorated by either genetic or pharmacological intervention reducing NOX activity, likely through reduced angII-mediated oxidative stress. Therefore, anti-NOX therapy merits evaluation to prevent the potentially serious cardiovascular complications of WBS, as well as in other cardiovascular disorders mediated by similar pathogenic mechanism.

    View details for DOI 10.1371/journal.pgen.1002458

    View details for Web of Science ID 000300725500002

    View details for PubMedID 22319452

  • Skeletogenic phenotype of human Marfan embryonic stem cells faithfully phenocopied by patient-specific induced-pluripotent stem cells PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Quarto, N., Leonard, B., Li, S., Marchand, M., Anderson, E., Behr, B., Francke, U., Reijo-Pera, R., Chiao, E., Longaker, M. T. 2012; 109 (1): 215-220

    Abstract

    Marfan syndrome (MFS) is a heritable connective tissue disorder caused by mutations in the gene coding for FIBRILLIN-1 (FBN1), an extracellular matrix protein. MFS is inherited as an autosomal dominant trait and displays major manifestations in the ocular, skeletal, and cardiovascular systems. Here we report molecular and phenotypic profiles of skeletogenesis in tissues differentiated from human embryonic stem cells and induced pluripotent stem cells that carry a heritable mutation in FBN1. We demonstrate that, as a biological consequence of the activation of TGF-β signaling, osteogenic differentiation of embryonic stem cells with a FBN1 mutation is inhibited; osteogenesis is rescued by inhibition of TGF-β signaling. In contrast, chondrogenesis is not perturbated and occurs in a TGF-β cell-autonomous fashion. Importantly, skeletal phenotypes observed in human embryonic stem cells carrying the monogenic FBN1 mutation (MFS cells) are faithfully phenocopied by cells differentiated from induced pluripotent-stem cells derived independently from MFS patient fibroblasts. Results indicate a unique phenotype uncovered by examination of mutant pluripotent stem cells and further demonstrate the faithful alignment of phenotypes in differentiated cells obtained from both human embryonic stem cells and induced pluripotent-stem cells, providing complementary and powerful tools to gain further insights into human molecular pathogenesis, especially of MFS.

    View details for DOI 10.1073/pnas.1113442109

    View details for Web of Science ID 000298876500045

    View details for PubMedID 22178754

    View details for PubMedCentralID PMC3252902

  • Efficient Replication of over 180 Genetic Associations with Self-Reported Medical Data PLOS ONE Tung, J. Y., Do, C. B., Hinds, D. A., Kiefer, A. K., Macpherson, J. M., Chowdry, A. B., Francke, U., Naughton, B. T., Mountain, J. L., Wojcicki, A., Eriksson, N. 2011; 6 (8)

    Abstract

    While the cost and speed of generating genomic data have come down dramatically in recent years, the slow pace of collecting medical data for large cohorts continues to hamper genetic research. Here we evaluate a novel online framework for obtaining large amounts of medical information from a recontactable cohort by assessing our ability to replicate genetic associations using these data. Using web-based questionnaires, we gathered self-reported data on 50 medical phenotypes from a generally unselected cohort of over 20,000 genotyped individuals. Of a list of genetic associations curated by NHGRI, we successfully replicated about 75% of the associations that we expected to (based on the number of cases in our cohort and reported odds ratios, and excluding a set of associations with contradictory published evidence). Altogether we replicated over 180 previously reported associations, including many for type 2 diabetes, prostate cancer, cholesterol levels, and multiple sclerosis. We found significant variation across categories of conditions in the percentage of expected associations that we were able to replicate, which may reflect systematic inflation of the effects in some initial reports, or differences across diseases in the likelihood of misdiagnosis or misreport. We also demonstrated that we could improve replication success by taking advantage of our recontactable cohort, offering more in-depth questions to refine self-reported diagnoses. Our data suggest that online collection of self-reported data from a recontactable cohort may be a viable method for both broad and deep phenotyping in large populations.

    View details for DOI 10.1371/journal.pone.0023473

    View details for Web of Science ID 000294121300051

    View details for PubMedID 21858135

    View details for PubMedCentralID PMC3157390

  • Web-Based Genome-Wide Association Study Identifies Two Novel Loci and a Substantial Genetic Component for Parkinson's Disease PLOS GENETICS Do, C. B., Tung, J. Y., Dorfman, E., Kiefer, A. K., Drabant, E. M., Francke, U., Mountain, J. L., Goldman, S. M., Tanner, C. M., Langston, J. W., Wojcicki, A., Eriksson, N. 2011; 7 (6)

    Abstract

    Although the causes of Parkinson's disease (PD) are thought to be primarily environmental, recent studies suggest that a number of genes influence susceptibility. Using targeted case recruitment and online survey instruments, we conducted the largest case-control genome-wide association study (GWAS) of PD based on a single collection of individuals to date (3,426 cases and 29,624 controls). We discovered two novel, genome-wide significant associations with PD-rs6812193 near SCARB2 (p = 7.6 × 10(-10), OR = 0.84) and rs11868035 near SREBF1/RAI1 (p = 5.6 × 10(-8), OR = 0.85)-both replicated in an independent cohort. We also replicated 20 previously discovered genetic associations (including LRRK2, GBA, SNCA, MAPT, GAK, and the HLA region), providing support for our novel study design. Relying on a recently proposed method based on genome-wide sharing estimates between distantly related individuals, we estimated the heritability of PD to be at least 0.27. Finally, using sparse regression techniques, we constructed predictive models that account for 6%-7% of the total variance in liability and that suggest the presence of true associations just beyond genome-wide significance, as confirmed through both internal and external cross-validation. These results indicate a substantial, but by no means total, contribution of genetics underlying susceptibility to both early-onset and late-onset PD, suggesting that, despite the novel associations discovered here and elsewhere, the majority of the genetic component for Parkinson's disease remains to be discovered.

    View details for DOI 10.1371/journal.pgen.1002141

    View details for Web of Science ID 000292386300054

    View details for PubMedID 21738487

  • Control of bone formation by the serpentine receptor Frizzled-9 JOURNAL OF CELL BIOLOGY Albers, J., Schulze, J., Beil, F. T., Gebauer, M., Baranowsky, A., Keller, J., Marshall, R. P., Wintges, K., Friedrich, F. W., Priemel, M., Schilling, A. F., Rueger, J. M., Cornils, K., Fehse, B., Streichert, T., Sauter, G., Jakob, F., Insogna, K. L., Pober, B., Knobeloch, K., Francke, U., Amling, M., Schinke, T. 2011; 192 (6): 1057-1072

    Abstract

    Although Wnt signaling in osteoblasts is of critical importance for the regulation of bone remodeling, it is not yet known which specific Wnt receptors of the Frizzled family are functionally relevant in this process. In this paper, we show that Fzd9 is induced upon osteoblast differentiation and that Fzd9(-/-) mice display low bone mass caused by impaired bone formation. Our analysis of Fzd9(-/-) primary osteoblasts demonstrated defects in matrix mineralization in spite of normal expression of established differentiation markers. In contrast, we observed a reduced expression of chemokines and interferon-regulated genes in Fzd9(-/-) osteoblasts. We also identified the ubiquitin-like modifier Isg15 as one potential downstream mediator of Fzd9 in these cells. Importantly, our molecular analysis further revealed that canonical Wnt signaling is not impaired in the absence of Fzd9, thus explaining the absence of a bone resorption phenotype. Collectively, our results reveal a previously unknown function of Fzd9 in osteoblasts, a finding that may have therapeutic implications for bone loss disorders.

    View details for DOI 10.1083/jcb.201008012

    View details for Web of Science ID 000288986200015

    View details for PubMedID 21402791

    View details for PubMedCentralID PMC3063134

  • Prognosis Factors in Probands With an FBN1 Mutation Diagnosed Before the Age of 1 Year PEDIATRIC RESEARCH Stheneur, C., Faivre, L., Collod-Beroud, G., Gautier, E., Binquet, C., Bonithon-Kopp, C., Claustres, M., Child, A. H., Arbustini, E., Ades, L. C., Francke, U., Mayer, K., Arslan-Kirchner, M., De Paepe, A., Chevallier, B., Bonnet, D., Jondeau, G., Boileau, C. 2011; 69 (3): 265-270

    Abstract

    Marfan syndrome (MFS) is an autosomal dominant connective tissue disorder. Diagnostic criteria of neonatal MFS (nMFS), the most severe form, are still debated. The aim of our study was to search for clinical and molecular prognostic factors that could be associated with length of survival. Probands ascertained via the framework of the Universal Marfan database-FBN1, diagnosed before the age of 1 y and presenting with cardiovascular features (aortic root dilatation or valvular insufficiency) were included in this study. Clinical and molecular data were correlated to survival. Among the 60 individuals, 38 had died, 82% died before the age of 1 y, mostly because of congestive heart failure. Three probands reached adulthood. Valvular insufficiencies and diaphragmatic hernia were predictive of shorter life expectancy. Two FBN1 mutations were found outside of the exon 24-32 region (in exons 4 and 21). Mutations in exons 25-26 were overrepresented and were associated with shorter survival (p = 0.03). We report the largest genotyped series of probands with MFS diagnosed before 1 y of life. In this population, factors significantly associated with shorter survival are presence of valvular insufficiencies or diaphragmatic hernia in addition to a mutation in exons 25 or 26.

    View details for Web of Science ID 000287621700015

    View details for PubMedID 21135753

  • Induced Chromosome Deletion in a Williams-Beuren Syndrome Mouse Model Causes Cardiovascular Abnormalities JOURNAL OF VASCULAR RESEARCH Goergen, C. J., Li, H., Francke, U., Taylor, C. A. 2011; 48 (2): 119-129

    Abstract

    The Williams-Beuren syndrome (WBS) is a genetic disorder caused by a heterozygous ~1.5-Mb deletion. The aim of this study was to determine how the genetic changes in a Wbs mouse model alter Eln expression, blood pressure, vessel structure, and abdominal aortic wall dynamics in vivo.Elastin (ELN) transcript levels were quantified by qRT-PCR and blood pressure was measured with a tail cuff system. M-mode ultrasound was used to track pulsatile abdominal aortic wall motion. Aortas were sectioned and stained to determine medial lamellar structure.ELN transcript levels were reduced by 38-41% in Wbs mice lacking one copy of the ELN gene. These mice also had a 10-20% increase in mean blood pressure and significantly reduced circumferential cyclic strain (p < 0.001). Finally, histological sections showed disorganized and fragmented elastin sheets in Wbs mice, but not the characteristic increase in lamellar units seen in Eln(+/-) mice.The deletion of Eln in this Wbs mouse model results in lower gene expression, hypertension, reduced cyclic strain, and fragmented elastin sheets. The observation that the number of medial lamellar units is normal in Wbs deletion mice, which is in contrast to Eln(+/-) mice, suggests other genes may be involved in vascular development.

    View details for DOI 10.1159/000316808

    View details for Web of Science ID 000282751600003

    View details for PubMedID 20926892

    View details for PubMedCentralID PMC2975735

  • Cardiovascular manifestations in men and women carrying a FBN1 mutation EUROPEAN HEART JOURNAL Detaint, D., Faivre, L., Collod-Beroud, G., Child, A. H., Loeys, B. L., Binquet, C., Gautier, E., Arbustini, E., Mayer, K., Arslan-Kirchner, M., Stheneur, C., Halliday, D., Beroud, C., Bonithon-Kopp, C., Claustres, M., Plauchu, H., Robinson, P. N., Kiotsekoglou, A., De Backer, J., Ades, L., Francke, U., De Paepe, A., Boileau, C., Jondeau, G. 2010; 31 (18): 2223-2229

    Abstract

    In patients with Marfan syndrome and other type-1 fibrillinopathies, genetic testing is becoming more easily available, leading to the identification of mutations early in the course of the disease. This study evaluates the cardiovascular (CV) risk associated with the discovery of a fibrillin-1 (FBN1) mutation.A total of 1,013 probands with pathogenic FBN1 mutations were included, among whom 965 patients [median age: 22 years (11-34), male gender 53%] had data suitable for analysis. The percentage of patients with an ascending aortic (AA) dilatation increased steadily with increasing age and reached 96% (95% CI: 94-97%) by 60 years. The presence of aortic events (dissection or prophylactic surgery) was rare before 20 years and then increased progressively, reaching 74% (95% CI: 67-81%) by 60 years. Compared with women, men were at higher risk for AA dilatation [≤ 30 years: 57% (95% CI: 52-63) vs. 50% (95% CI: 45-55), P = 0.0076] and aortic events [≤ 30 years: 21% (95% CI: 17-26) vs. 11% (95% CI: 8-16), P < 0.0001; adjusted HR: 1.4 (1.1-1.8), P = 0.005]. The prevalence of mitral valve (MV) prolapse [≤ 60 years: 77% (95% CI: 72-82)] and MV regurgitation [≤ 60 years: 61% (95% CI: 53-69)] also increased steadily with age, but surgery limited to the MV remained rare [≤ 60 years: 13% (95% CI: 8-21)]. No difference between genders was observed (for all P> 0.20). From 1985 to 2005 the prevalence of AA dilatation remained stable (P for trend = 0.88), whereas the percentage of patients with AA dissection significantly decreased (P for trend = 0.01).The CV risk remains important in patients with an FBN1 gene mutation and is present throughout life, justifying regular aortic monitoring. Aortic dilatation or dissection should always trigger suspicion of a genetic background leading to thorough examination for extra-aortic features and comprehensive pedigree investigation.

    View details for DOI 10.1093/eurheartj/ehq258

    View details for Web of Science ID 000281955900012

    View details for PubMedID 20709720

  • Neonatal Maternal Deprivation Response and Developmental Changes in Gene Expression Revealed by Hypothalamic Gene Expression Profiling in Mice PLOS ONE Ding, F., Li, H. H., Li, J., Myers, R. M., Francke, U. 2010; 5 (2)

    Abstract

    Neonatal feeding problems are observed in several genetic diseases including Prader-Willi syndrome (PWS). Later in life, individuals with PWS develop hyperphagia and obesity due to lack of appetite control. We hypothesized that failure to thrive in infancy and later-onset hyperphagia are related and could be due to a defect in the hypothalamus. In this study, we performed gene expression microarray analysis of the hypothalamic response to maternal deprivation in neonatal wild-type and Snord116del mice, a mouse model for PWS in which a cluster of imprinted C/D box snoRNAs is deleted. The neonatal starvation response in both strains was dramatically different from that reported in adult rodents. Genes that are affected by adult starvation showed no expression change in the hypothalamus of 5 day-old pups after 6 hours of maternal deprivation. Unlike in adult rodents, expression levels of Nanos2 and Pdk4 were increased, and those of Pgpep1, Ndp, Brms1l, Mett10d, and Snx1 were decreased after neonatal deprivation. In addition, we compared hypothalamic gene expression profiles at postnatal days 5 and 13 and observed significant developmental changes. Notably, the gene expression profiles of Snord116del deletion mice and wild-type littermates were very similar at all time points and conditions, arguing against a role of Snord116 in feeding regulation in the neonatal period.

    View details for DOI 10.1371/journal.pone.0009402

    View details for Web of Science ID 000274924200023

    View details for PubMedID 20195375

    View details for PubMedCentralID PMC2827556

  • On the bumpy road towards 'personalized medicine' EMBO MOLECULAR MEDICINE Francke, U. 2010; 2 (1): 1-2

    View details for DOI 10.1002/emmm.200900056

    View details for Web of Science ID 000274291000001

    View details for PubMedID 20043276

  • Widespread Changes in Dendritic and Axonal Morphology in Mecp2-Mutant Mouse Models of Rett Syndrome: Evidence for Disruption of Neuronal Networks JOURNAL OF COMPARATIVE NEUROLOGY Belichenko, P. V., Wright, E. E., Belichenko, N. P., Masliah, E., Li, H. H., Mobley, W. C., Francke, U. 2009; 514 (3): 240-258

    Abstract

    Rett syndrome (RTT) is a neurodevelopmental disorder caused by mutations in the X-linked gene MECP2. Girls with RTT show dramatic changes in brain function, but relatively few studies have explored the structure of neural circuits. Examining two mouse models of RTT (Mecp2B and Mecp2J), we previously documented changes in brain anatomy. Herein, we use confocal microscopy to study the effects of MeCP2 deficiency on the morphology of dendrites and axons in the fascia dentata (FD), CA1 area of hippocampus, and motor cortex following Lucifer yellow microinjection or carbocyanine dye tracing. At 3 weeks of age, most (33 of 41) morphological parameters were significantly altered in Mecp2B mice; fewer (23 of 39) were abnormal in Mecp2J mice. There were striking changes in the density and size of the dendritic spines and density and orientation of axons. In Mecp2B mice, dendritic spine density was decreased in the FD (approximately 11%), CA1 (14-22%), and motor cortex (approximately 16%). A decreased spine head size (approximately 9%) and an increased spine neck length (approximately 12%) were found in Mecp2B FD. In addition, axons in the motor cortex were disorganized. In Mecp2J mice, spine density was significantly decreased in CA1 (14-26%). In both models, dendritic swelling and elongated spine necks were seen in all areas studied. Marked variation in the type and extent of changes was noted in dendrites of adjacent neurons. Electron microscopy confirmed abnormalities in dendrites and axons and showed abnormal mitochondria. Our findings document widespread abnormalities of dendrites and axons that recapitulate those seen in RTT.

    View details for DOI 10.1002/cne.22009

    View details for Web of Science ID 000264907800003

    View details for PubMedID 19296534

  • Pathogenic FBN1 Mutations in 146 Adults Not Meeting Clinical Diagnostic Criteria for Marfan Syndrome: Further Delineation of Type 1 Fibrillinopathies and Focus on Patients With an Isolated Major Criterion AMERICAN JOURNAL OF MEDICAL GENETICS PART A Faivre, L., Collod-Beroud, G., Callewaert, B., Child, A., Loeys, B. L., Binquet, C., Gautier, E., Arbustini, E., Mayer, K., Arslan-Kirchner, M., Kiotsekoglou, A., Comeglio, P., Grasso, M., Beroud, C., Bonithon-Kopp, C., Claustres, M., Stheneur, C., Bouchot, O., Wolf, J. E., Robinson, P. N., Ades, L., De Backer, J., Coucke, P., FRANCKE, U., De Paepe, A., Boileau, C., Jondeau, G. 2009; 149A (5): 854-860

    Abstract

    Mutations in the FBN1 gene cause Marfan syndrome (MFS) and have been associated with a wide range of milder overlapping phenotypes. A proportion of patients carrying a FBN1 mutation does not meet diagnostic criteria for MFS, and are diagnosed with "other type I fibrillinopathy." In order to better describe this entity, we analyzed a subgroup of 146 out of 689 adult propositi with incomplete "clinical" international criteria (Ghent nosology) from a large collaborative international study including 1,009 propositi with a pathogenic FBN1 mutation. We focused on patients with only one major clinical criterion, [including isolated ectopia lentis (EL; 12 patients), isolated ascending aortic dilatation (17 patients), and isolated major skeletal manifestations (1 patient)] or with no major criterion but only minor criteria in 1 or more organ systems (16 patients). At least one component of the Ghent nosology, insufficient alone to make a minor criterion, was found in the majority of patients with isolated ascending aortic dilatation and isolated EL. In patients with isolated EL, missense mutations involving a cysteine were predominant, mutations in exons 24-32 were underrepresented, and no mutations leading to a premature truncation were found. Studies of recurrent mutations and affected family members of propositi with only one major clinical criterion argue for a clinical continuum between such phenotypes and classical MFS. Using strict definitions, we conclude that patients with FBN1 mutation and only one major clinical criterion or with only minor clinical criteria of one or more organ system do exist but represent only 5% of the adult cohort.

    View details for DOI 10.1002/ajmg.a.32809

    View details for Web of Science ID 000265805900007

    View details for PubMedID 19353630

  • Clinical and mutation-type analysis from an international series of 198 probands with a pathogenic FBN1 exons 24-32 mutation EUROPEAN JOURNAL OF HUMAN GENETICS Faivre, L., Collod-Beroud, G., Callewaert, B., Child, A., Binquet, C., Gautier, E., Loeys, B. L., Arbustini, E., Mayer, K., Arslan-Kirchner, M., Stheneur, C., Kiotsekoglou, A., Comeglio, P., Marziliano, N., Wolf, J. E., Bouchot, O., Khau-Van-Kien, P., Beroud, C., Claustres, M., Bonithon-Kopp, C., Robinson, P. N., Ades, L., De Backer, J., Coucke, P., FRANCKE, U., De Paepe, A., Jondeau, G., Boileau, C. 2009; 17 (4): 491-501

    Abstract

    Mutations in the FBN1 gene cause Marfan syndrome (MFS) and a wide range of overlapping phenotypes. The severe end of the spectrum is represented by neonatal MFS, the vast majority of probands carrying a mutation within exons 24-32. We previously showed that a mutation in exons 24-32 is predictive of a severe cardiovascular phenotype even in non-neonatal cases, and that mutations leading to premature truncation codons are under-represented in this region. To describe patients carrying a mutation in this so-called 'neonatal' region, we studied the clinical and molecular characteristics of 198 probands with a mutation in exons 24-32 from a series of 1013 probands with a FBN1 mutation (20%). When comparing patients with mutations leading to a premature termination codon (PTC) within exons 24-32 to patients with an in-frame mutation within the same region, a significantly higher probability of developing ectopia lentis and mitral insufficiency were found in the second group. Patients with a PTC within exons 24-32 rarely displayed a neonatal or severe MFS presentation. We also found a higher probability of neonatal presentations associated with exon 25 mutations, as well as a higher probability of cardiovascular manifestations. A high phenotypic heterogeneity could be described for recurrent mutations, ranging from neonatal to classical MFS phenotype. In conclusion, even if the exons 24-32 location appears as a major cause of the severity of the phenotype in patients with a mutation in this region, other factors such as the type of mutation or modifier genes might also be relevant.

    View details for DOI 10.1038/ejhg.2008.207

    View details for Web of Science ID 000264354900012

    View details for PubMedID 19002209

  • Induced chromosome deletions cause hypersociability and other features of Williams-Beuren syndrome in mice EMBO MOLECULAR MEDICINE Li, H. H., Roy, M., Kuscuoglu, U., Spencer, C. M., Halm, B., Harrison, K. C., Bayle, J. H., Splendore, A., Ding, F., Meltzer, L. A., Wright, E., Paylor, R., Deisseroth, K., Francke, U. 2009; 1 (1): 50-65

    Abstract

    The neurodevelopmental disorder Williams-Beuren syndrome is caused by spontaneous approximately 1.5 Mb deletions comprising 25 genes on human chromosome 7q11.23. To functionally dissect the deletion and identify dosage-sensitive genes, we created two half-deletions of the conserved syntenic region on mouse chromosome 5G2. Proximal deletion (PD) mice lack Gtf2i to Limk1, distal deletion (DD) mice lack Limk1 to Fkbp6, and the double heterozygotes (D/P) model the complete human deletion. Gene transcript levels in brain are generally consistent with gene dosage. Increased sociability and acoustic startle response are associated with PD, and cognitive defects with DD. Both PD and D/P males are growth-retarded, while skulls are shortened and brains are smaller in DD and D/P. Lateral ventricle (LV) volumes are reduced, and neuronal cell density in the somatosensory cortex is increased, in PD and D/P. Motor skills are most impaired in D/P. Together, these partial deletion mice replicate crucial aspects of the human disorder and serve to identify genes and gene networks contributing to the neural substrates of complex behaviours and behavioural disorders.

    View details for DOI 10.1002/emmm.200900003

    View details for Web of Science ID 000273437400010

    View details for PubMedID 20049703

    View details for PubMedCentralID PMC3378107

  • Clinical and Molecular Study of 320 Children With Marfan Syndrome and Related Type I Fibrillinopathies in a Series of 1009 Probands With Pathogenic FBN1 Mutations PEDIATRICS Faivre, L., Masurel-Paulet, A., Collod-Beroud, G., Callewaert, B. L., Child, A. H., Stheneur, C., Binquet, C., Gautier, E., Chevallier, B., Huet, F., Loeys, B. L., Arbustini, E., Mayer, K., Arslan-Kirchner, M., Kiotsekoglou, A., Comeglio, P., Grasso, M., Halliday, D. J., Beroud, C., Bonithon-Kopp, C., Claustres, M., Robinson, P. N., Ades, L., De Backer, J., Coucke, P., Francke, U., De Paepe, A., Boileau, C., Jondeau, G. 2009; 123 (1): 391-398

    Abstract

    From a large series of 1009 probands with pathogenic FBN1 mutations, data for 320 patients <18 years of age at the last follow-up evaluation were analyzed (32%). At the time of diagnosis, the median age was 6.5 years. At the last examination, the population was classified as follows: neonatal Marfan syndrome, 14%; severe Marfan syndrome, 19%; classic Marfan syndrome, 32%; probable Marfan syndrome, 35%. Seventy-one percent had ascending aortic dilation, 55% ectopia lentis, and 28% major skeletal system involvement. Even when aortic complications existed in childhood, the rates of aortic surgery and aortic dissection remained low (5% and 1%, respectively). Some diagnostic features (major skeletal system involvement, striae, dural ectasia, and family history) were more frequent in the 10- to <18-year age group, whereas others (ascending aortic dilation and mitral abnormalities) were more frequent in the population with neonatal Marfan syndrome. Only 56% of children could be classified as having Marfan syndrome, according to international criteria, at their last follow-up evaluation when the presence of a FBN1 mutation was not considered as a major feature, with increasing frequency in the older age groups. Eighty-five percent of child probands fulfilled international criteria after molecular studies, which indicates that the discovery of a FBN1 mutation can be a valuable diagnostic aid in uncertain cases. The distributions of mutation types and locations in this pediatric series revealed large proportions of probands carrying mutations located in exons 24 to 32 (33%) and in-frame mutations (75%). Apart from lethal neonatal Marfan syndrome, we confirm that the majority of clinical manifestations of Marfan syndrome increase with age, which emphasizes the poor applicability of the international criteria to this diagnosis in childhood and the need for follow-up monitoring in cases of clinical suspicion of Marfan syndrome.

    View details for DOI 10.1542/peds.2008-0703

    View details for Web of Science ID 000262046400057

    View details for PubMedID 19117906

  • Severe congenital encephalopathy caused by MECP2 null mutations in males: central hypoxia and reduced neuronal dendritic structure CLINICAL GENETICS Schule, B., Armstrong, D. D., Vogel, H., Oviedo, A., Francke, U. 2008; 74 (2): 116-126

    Abstract

    Non-mosaic males with a 46,XY karyotype and a MECP2 null mutation display a phenotype of severe neonatal-onset encephalopathy that is distinctly different from Rett syndrome (RTT). To increase awareness of this rare disorder, we are reporting novel findings in a sporadic case, compare them to 16 previously reported cases and establish salient criteria for clinical diagnosis. The proband suffered from general hypotonia and hypoxia caused by hypoventilation and irregular breathing. He developed abnormal movements, seizures and electroencephalogram abnormalities. He failed to thrive and to reach any motor milestones and died at 15 months from central respiratory failure without a diagnosis. In a muscle biopsy, type II fibers were reduced in diameter, indicating central hypoxia. At autopsy, the brain was small with disproportionate reduction of the frontal and temporal lobes. Synaptophysin staining of synaptic vesicles was greatly reduced in cerebellar and spinal cord sections. Analysis of Golgi-stained pyramidal neurons from cortical layers III and V of the frontal and temporal lobes revealed drastically diminished dendritic trees. Post-mortem MECP2 mutation analysis on DNA and RNA from fibroblasts revealed a novel de novo 9-nucleotide deletion including the intron 3/exon 4 splice junction. The two nucleotides flanking the deletion form a new splice site, and the aberrantly spliced transcript lacks seven nucleotides (r.378_384delTCCCCAG), causing a frameshift and premature termination codon (p.I126fsX11). Males with congenital encephalopathy, not females with RTT, represent the true human counterpart for the commonly studied Mecp2-/y mouse model and provide unique insight into the mechanisms of MeCP2 deficiency.

    View details for DOI 10.1111/j.1399-0004.2008.01005.x

    View details for Web of Science ID 000257476200003

    View details for PubMedID 18477000

  • Contribution of molecular analyses in diagnosing Marfan syndrome and type I fibrillinopathies: an international study of 1009 probands JOURNAL OF MEDICAL GENETICS Faivre, L., Collod-Beroud, G., Child, A., Callewaert, B., Loeys, B. L., Binquet, C., Gautier, E., Arbustini, E., Mayer, K., Arslan-Kirchner, M., Stheneur, C., Kiotsekoglou, A., Comeglio, P., Marziliano, N., Halliday, D., Beroud, C., Bonithon-Kopp, C., Claustres, M., Plauchu, H., Robinson, P. N., Ades, L., De Backer, J., Coucke, P., FRANCKE, U., De Paepe, A., Boileau, C., Jondeau, G. 2008; 45 (6): 384-390

    Abstract

    The diagnosis of Marfan syndrome (MFS) is usually initially based on clinical criteria according to the number of major and minor systems affected following international nosology. The number of FBN1 mutation carriers, at risk of aortic complications who would not be properly diagnosed based only on clinical grounds, is of growing importance owing to the increased availability of molecular screening. The aim of the study was to identify patients who should be considered for FBN1 mutation screening.Our international series included 1009 probands with a known FBN1 mutation. Patients were classified as either fulfilling or not fulfilling "clinical" criteria. In patients with unfulfilled "clinical" criteria, we evaluated the percentage of additional patients who became positive for international criteria when the FBN1 mutation was considered. The aortic risk was evaluated and compared in patients fulfilling or not fulfilling the "clinical" international criteria.Diagnosis of MFS was possible on clinical grounds in 79% of the adults, whereas 90% fulfilled the international criteria when including the FBN1 mutation. Corresponding figures for children were 56% and 85%, respectively. Aortic dilatation occurred later in adults with unfulfilled "clinical criteria" when compared to the Marfan syndrome group (44% vs 73% at 40 years, p<0.001), but the lifelong risk for ascending aortic dissection or surgery was not significantly different in both groups.Because of its implications for aortic follow-up, FBN1 molecular analysis is recommended in newly suspected MFS when two systems are involved with at least one major system affected. This is of utmost importance in patients without aortic dilatation and in children.

    View details for DOI 10.1136/jmg.2007.056382

    View details for Web of Science ID 000256369500009

    View details for PubMedID 18310266

  • Comparative study of brain morphology in Mecp2 mutant mouse models of Rett syndrome JOURNAL OF COMPARATIVE NEUROLOGY Belichenko, N. P., Belichenko, P. V., Li, H. H., Mobley, W. C., Francke, U. 2008; 508 (1): 184-195

    Abstract

    Rett syndrome (RTT) is caused by mutations in the X-linked gene MECP2. While patients with RTT show widespread changes in brain function, relatively few studies document changes in brain structure and none examine in detail whether mutations causing more severe clinical phenotypes are linked to more marked changes in brain structure. To study the influence of MeCP2-deficiency on the morphology of brain areas and axonal bundles, we carried out an extensive morphometric study of two Mecp2-mutant mouse models (Mecp2B and Mecp2J) of RTT. Compared to wildtype littermates, striking changes included reduced brain weight ( approximately 13% and approximately 9%) and the volumes of cortex ( approximately 11% and approximately 7%), hippocampus (both by approximately 8%), and cerebellum ( approximately 12% and 8%) in both mutant mice. At 3 weeks of age, most (24 of 47) morphological parameters were significantly altered in Mecp2B mice; fewer (18) were abnormal in Mecp2J mice. In Mecp2B mice, significantly lower values for cortical area were distributed along the rostrocaudal axis, and there was a reduced length of the olfactory bulb ( approximately 10%) and periaqueductal gray matter ( approximately 16%). In Mecp2J mice, while there was significant reduction in rostrocaudal length of cortex, this parameter was also abnormal in hippocampus ( approximately 10%), periaqueductal gray matter ( approximately 13%), fimbria ( approximately 18%), and anterior commissure ( approximately 10%). Our findings define patterns of Mecp2 mutation-induced changes in brain structure that are widespread and show that while some changes are present in both mutants, others are not. These observations provide the underpinning for studies to further define microarchitectural and physiological consequences of MECP2 deficiency.

    View details for DOI 10.1002/cne.21673

    View details for Web of Science ID 000254709900012

    View details for PubMedID 18306326

  • SnoRNA Snord116 (Pwcr1/MBII-85) Deletion Causes Growth Deficiency and Hyperphagia in Mice PLOS ONE Ding, F., Li, H. H., Zhang, S., Solomon, N. M., Camper, S. A., Cohen, P., Francke, U. 2008; 3 (3)

    Abstract

    Prader-Willi syndrome (PWS) is the leading genetic cause of obesity. After initial severe hypotonia, PWS children become hyperphagic and morbidly obese, if intake is not restricted. Short stature with abnormal growth hormone secretion, hypogonadism, cognitive impairment, anxiety and behavior problems are other features. PWS is caused by lack of expression of imprinted genes in a approximately 4 mb region of chromosome band 15q11.2. Our previous translocation studies predicted a major role for the C/D box small nucleolar RNA cluster SNORD116 (PWCR1/HBII-85) in PWS. To test this hypothesis, we created a approximately 150 kb deletion of the > 40 copies of Snord116 (Pwcr1/MBII-85) in C57BL/6 mice. Snord116del mice with paternally derived deletion lack expression of this snoRNA. They have early-onset postnatal growth deficiency, but normal fertility and lifespan. While pituitary structure and somatotrophs are normal, liver Igf1 mRNA is decreased. In cognitive and behavior tests, Snord116del mice are deficient in motor learning and have increased anxiety. Around three months of age, they develop hyperphagia, but stay lean on regular and high-fat diet. On reduced caloric intake, Snord116del mice maintain their weight better than wild-type littermates, excluding increased energy requirement as a cause of hyperphagia. Normal compensatory feeding after fasting, and ability to maintain body temperature in the cold indicate normal energy homeostasis regulation. Metabolic chamber studies reveal that Snord116del mice maintain energy homeostasis by altered fuel usage. Prolonged mealtime and increased circulating ghrelin indicate a defect in meal termination mechanism. Snord116del mice, the first snoRNA deletion animal model, reveal a novel role for a non-coding RNA in growth and feeding regulation.

    View details for DOI 10.1371/journal.pone.0001709

    View details for Web of Science ID 000260586600005

    View details for PubMedID 18320030

    View details for PubMedCentralID PMC2248623

  • Reduced aortic wall motion and cyclic strain in a Williams-Beuren syndrome mouse model due to changes in vessel structure 80th Annual Scientific Session of the American-Heart-Association (AHA) Goergen, C. J., Li, H. H., Abilez, O. J., Francke, U., Taylor, C. A. LIPPINCOTT WILLIAMS & WILKINS. 2007: 33–33
  • A Marfan syndrome gene expression phenotype in cultured skin fibroblasts BMC GENOMICS Yao, Z., Jaeger, J. C., Ruzzo, W. L., Morale, C. Z., Emond, M., Francke, U., Milewicz, D. M., Schwartz, S. M., Mulvihill, E. R. 2007; 8

    Abstract

    Marfan syndrome (MFS) is a heritable connective tissue disorder caused by mutations in the fibrillin-1 gene. This syndrome constitutes a significant identifiable subtype of aortic aneurysmal disease, accounting for over 5% of ascending and thoracic aortic aneurysms.We used spotted membrane DNA macroarrays to identify genes whose altered expression levels may contribute to the phenotype of the disease. Our analysis of 4132 genes identified a subset with significant expression differences between skin fibroblast cultures from unaffected controls versus cultures from affected individuals with known fibrillin-1 mutations. Subsequently, 10 genes were chosen for validation by quantitative RT-PCR.Differential expression of many of the validated genes was associated with MFS samples when an additional group of unaffected and MFS affected subjects were analyzed (p-value < 3 x 10-6 under the null hypothesis that expression levels in cultured fibroblasts are unaffected by MFS status). An unexpected observation was the range of individual gene expression. In unaffected control subjects, expression ranges exceeding 10 fold were seen in many of the genes selected for qRT-PCR validation. The variation in expression in the MFS affected subjects was even greater.

    View details for DOI 10.1186/1471-2164-8-319

    View details for Web of Science ID 000252437300001

    View details for PubMedID 17850668

  • DLX5 and DLX6 expression is biallelic and not modulated by MeCP2 deficiency AMERICAN JOURNAL OF HUMAN GENETICS Schuele, B., Li, H. H., Fisch-Kohl, C., Purmann, C., Francke, U. 2007; 81 (3): 492-506

    Abstract

    Mutations in MECP2 and Mecp2 (encoding methyl-CpG binding protein 2 [MeCP2]) cause distinct neurological phenotypes in humans and mice, respectively, but the molecular pathology is unclear. Recent literature claimed that the developmental homeobox gene DLX5 is imprinted and that its imprinting status is modulated by MeCP2, leading to biallelic expression in Rett syndrome and twofold overexpression of Dlx5 and Dlx6 in Mecp2-null mice. The conclusion that DLX5 is a direct target of MeCP2 has implications for research on the molecular bases of Rett syndrome, autism, and genomic imprinting. Attempting to replicate the reported data, we evaluated allele-specific expression of DLX5 and DLX6 in mouse x human somatic cell hybrids, lymphoblastoid cell lines, and frontal cortex from controls and individuals with MECP2 mutations. We identified novel single-nucleotide polymorphisms in DLX5 and DLX6, enabling the first imprinting studies of DLX6. We found that DLX5 and DLX6 are biallelically expressed in somatic cell hybrids and in human cell lines and brain, with no differences between affected and control samples. We also determined expression levels of Dlx5 and Dlx6 in forebrain from seven male Mecp2-mutant mice and eight wild-type littermates by real-time quantitative reverse-transcriptase polymerase chain reaction assays. Expression of Dlx5 and Dlx6, as well as of the imprinted gene Peg3, in mouse forebrain was highly variable, with no consistent differences between Mecp2-null mutants and controls. We conclude that DLX5 and DLX6 are not imprinted in humans and are not likely to be direct targets of MeCP2 modulation. In contrast, the imprinting status of PEG3 and PEG10 is maintained in MeCP2-deficient tissues. Our results confirm that MeCP2 plays no role in the maintenance of genomic imprinting and add PEG3 and PEG10 to the list of studied imprinted genes.

    View details for DOI 10.1086/520063

    View details for Web of Science ID 000249128200006

    View details for PubMedID 17701895

    View details for PubMedCentralID PMC1950824

  • Effect of mutation type and location on clinical outcome in 1,013 probands with Marfan syndrome or related phenotypes and FBN1 mutations: An international study AMERICAN JOURNAL OF HUMAN GENETICS Faivre, L., Collod-Beroud, G., Loeys, B. L., Child, A., Binquet, C., Gautier, E., Callewaert, B., Arbustini, E., Mayer, K., Arslan-Kirchner, M., Kiotsekoglou, A., Comeglio, P., Marziliano, N., Dietz, H. C., Halliday, D., Beroud, C., Bonithon-Kopp, C., Claustres, M., Muti, C., Plauchu, H., Robinson, P. N., Ades, L. C., Biggin, A., Benetts, B., Brett, M., Holman, K. J., De Backer, J., Coucke, P., FRANCKE, U., De Paepe, A., Jondeau, G., Boileau, C. 2007; 81 (3): 454-466

    Abstract

    Mutations in the fibrillin-1 (FBN1) gene cause Marfan syndrome (MFS) and have been associated with a wide range of overlapping phenotypes. Clinical care is complicated by variable age at onset and the wide range of severity of aortic features. The factors that modulate phenotypical severity, both among and within families, remain to be determined. The availability of international FBN1 mutation Universal Mutation Database (UMD-FBN1) has allowed us to perform the largest collaborative study ever reported, to investigate the correlation between the FBN1 genotype and the nature and severity of the clinical phenotype. A range of qualitative and quantitative clinical parameters (skeletal, cardiovascular, ophthalmologic, skin, pulmonary, and dural) was compared for different classes of mutation (types and locations) in 1,013 probands with a pathogenic FBN1 mutation. A higher probability of ectopia lentis was found for patients with a missense mutation substituting or producing a cysteine, when compared with other missense mutations. Patients with an FBN1 premature termination codon had a more severe skeletal and skin phenotype than did patients with an inframe mutation. Mutations in exons 24-32 were associated with a more severe and complete phenotype, including younger age at diagnosis of type I fibrillinopathy and higher probability of developing ectopia lentis, ascending aortic dilatation, aortic surgery, mitral valve abnormalities, scoliosis, and shorter survival; the majority of these results were replicated even when cases of neonatal MFS were excluded. These correlations, found between different mutation types and clinical manifestations, might be explained by different underlying genetic mechanisms (dominant negative versus haploinsufficiency) and by consideration of the two main physiological functions of fibrillin-1 (structural versus mediator of TGF beta signalling). Exon 24-32 mutations define a high-risk group for cardiac manifestations associated with severe prognosis at all ages.

    View details for DOI 10.1086/520125

    View details for Web of Science ID 000249128200003

    View details for PubMedID 17701892

  • Cerebellar gene expression profiles of mouse models for Rett syndrome reveal novel MeCP2 targets BMC MEDICAL GENETICS Jordan, C., Li, H. H., Kwan, H. C., Francke, U. 2007; 8

    Abstract

    MeCP2, methyl-CpG-binding protein 2, binds to methylated cytosines at CpG dinucleotides, as well as to unmethylated DNA, and affects chromatin condensation. MECP2 mutations in females lead to Rett syndrome, a neurological disorder characterized by developmental stagnation and regression, loss of purposeful hand movements and speech, stereotypic hand movements, deceleration of brain growth, autonomic dysfunction and seizures. Most mutations occur de novo during spermatogenesis. Located at Xq28, MECP2 is subject to X inactivation, and affected females are mosaic. Rare hemizygous males suffer from a severe congenital encephalopathy.To identify the pathways mis-regulated by MeCP2 deficiency, microarray-based global gene expression studies were carried out in cerebellum of Mecp2 mutant mice. We compared transcript levels in mutant/wildtype male sibs of two different MeCP2-deficient mouse models at 2, 4 and 8 weeks of age. Increased transcript levels were evaluated by real-time quantitative RT-PCR. Chromatin immunoprecipitation assays were used to document in vivo MeCP2 binding to promoter regions of candidate target genes.Of several hundred genes with altered expression levels in the mutants, twice as many were increased than decreased, and only 27 were differentially expressed at more than one time point. The number of misregulated genes was 30% lower in mice with the exon 3 deletion (Mecp2tm1.1Jae) than in mice with the larger deletion (Mecp2tm1.1Bird). Between the mutants, few genes overlapped at each time point. Real-time quantitative RT-PCR assays validated increased transcript levels for four genes: Irak1, interleukin-1 receptor-associated kinase 1; Fxyd1, phospholemman, associated with Na, K-ATPase;Reln, encoding an extracellular signaling molecule essential for neuronal lamination and synaptic plasticity; and Gtl2/Meg3, an imprinted maternally expressed non-translated RNA that serves as a host gene for C/D box snoRNAs and microRNAs. Chromatin immunoprecipitation assays documented in vivo MeCP2 binding to promoter regions of Fxyd1, Reln, and Gtl2.Transcriptional profiling of cerebellum failed to detect significant global changes in Mecp2-mutant mice. Increased transcript levels of Irak1, Fxyd1, Reln, and Gtl2 may contribute to the neuronal dysfunction in MeCP2-deficient mice and individuals with Rett syndrome. Our data provide testable hypotheses for future studies of the regulatory or signaling pathways that these genes act on.

    View details for DOI 10.1186/1471-2350-8-36

    View details for Web of Science ID 000248478800002

    View details for PubMedID 17584923

    View details for PubMedCentralID PMC1931432

  • An atypical deletion of the Williams-Beuren syndrome interval implicates genes associated with defective visuospatial processing and autism JOURNAL OF MEDICAL GENETICS Edelmann, L., Prosnitz, A., Pardo, S., Bhatt, J., Cohen, N., Lauriat, T., Ouchanov, L., Gonzalez, P. J., Manghi, E. R., Bondy, P., Esquivel, M., Monge, S., Delgado, M. F., Splendore, A., Francke, U., Burton, B. K., McInnes, L. A. 2007; 44 (2): 136-143

    Abstract

    During a genetic study of autism, a female child who met diagnostic criteria for autism spectrum disorder, but also exhibited the cognitive-behavioural profile (CBP) associated with Williams-Beuren syndrome (WBS) was examined. The WBS CBP includes impaired visuospatial ability, an overly friendly personality, excessive non-social anxiety and language delay.Using array-based comparative genomic hybridisation (aCGH), a deletion corresponding to BAC RP11-89A20 in the distal end of the WBS deletion interval was detected. Hemizygosity was confirmed using fluorescence in situ hybridisation and fine mapping was performed by measuring the copy number of genomic DNA using quantitative polymerase chain reaction.The proximal breakpoint was mapped to intron 1 of GTF2IRD1 and the distal breakpoint lies 2.4-3.1 Mb towards the telomere. The subject was completely hemizygous for GTF2I, commonly deleted in carriers of the classic approximately 1.5 Mb WBS deletion, and GTF2IRD2, deleted in carriers of the rare approximately 1.84 Mb WBS deletion.Hemizygosity of the GTF2 family of transcription factors is sufficient to produce many aspects of the WBS CBP, and particularly implicate the GTF2 transcription factors in the visuospatial construction deficit. Symptoms of autism in this case may be due to deletion of additional genes outside the typical WBS interval or remote effects on gene expression at other loci.

    View details for DOI 10.1136/jmg.2006.044537

    View details for Web of Science ID 000244098100007

    View details for PubMedID 16971481

    View details for PubMedCentralID PMC2598069

  • Ube3a expression is not altered in Mecp2 mutant mice HUMAN MOLECULAR GENETICS Jordan, C., Francke, U. 2006; 15 (14): 2210-2215

    Abstract

    Rett syndrome (RTT) is a neurodevelopmental disorder characterized by cognitive regression, loss of purposeful hand movements and speech, stereotypies, ataxia, seizures, mental retardation and acquired microcephaly. Mutations in MECP2, encoding methyl-CpG-binding protein 2, are responsible for approximately 90% of classic RTT cases. RTT displays phenotypic overlap with Angelman syndrome, a disorder caused by loss of expression of the imprinted gene UBE3A. MeCP2 binds to methylated DNA and may alter the expression of imprinted genes, thereby suggesting a mechanistic link between the two disorders. Here, we tested the hypothesis that MeCP2 deficiency affects expression of Ube3a in mouse models of RTT. As Ube3a is only imprinted in brain, we evaluated Ube3a expression in brains of 15 different litters of neonatal or 8-week-old male Mecp2 mutant mice by real-time quantitative RT-PCR and western blot analysis. We found no significant differences between Mecp2(tm1.1Bird/Y) or Mecp2(tm1.1Jae/Y) mutants and their wild-type male siblings that served as negative controls. In positive control mice carrying a maternally inherited Ube3a deletion, Ube3a sense transcript and protein levels were drastically reduced. Our data contrast with two recent reports of substantially decreased Ube3a expression in brain tissues of MeCP2-deficient mice. We, therefore, challenge the conclusion that decreased UBE3A/Ube3a expression contributes to the pathophysiology of RTT.

    View details for DOI 10.1093/hmg/ddl146

    View details for Web of Science ID 000238762800004

    View details for PubMedID 16754645

  • NFAT dysregulation by increased dosage of DSCR1 and DYRK1A on chromosome 21 NATURE Arron, J. R., Winslow, M. M., Polleri, A., Chang, C., Wu, H., Gao, X., Neilson, J. R., Chen, L., Heit, J. J., Kim, S. K., Yamasaki, N., Miyakawa, T., Francke, U., Graef, I. A., Crabtree, G. R. 2006; 441 (7093): 595-600

    Abstract

    Trisomy 21 results in Down's syndrome, but little is known about how a 1.5-fold increase in gene dosage produces the pleiotropic phenotypes of Down's syndrome. Here we report that two genes, DSCR1 and DYRK1A , lie within the critical region of human chromosome 21 and act synergistically to prevent nuclear occupancy of NFATc transcription factors, which are regulators of vertebrate development. We use mathematical modelling to predict that autoregulation within the pathway accentuates the effects of trisomy of DSCR1 and DYRK1A, leading to failure to activate NFATc target genes under specific conditions. Our observations of calcineurin-and Nfatc-deficient mice, Dscr1- and Dyrk1a-overexpressing mice, mouse models of Down's syndrome and human trisomy 21 are consistent with these predictions. We suggest that the 1.5-fold increase in dosage of DSCR1 and DYRK1A cooperatively destabilizes a regulatory circuit, leading to reduced NFATc activity and many of the features of Down's syndrome. More generally, these observations suggest that the destabilization of regulatory circuits can underlie human disease.

    View details for DOI 10.1038/nature04678

    View details for PubMedID 16554754

  • Identification of cis-regulatory elements for MECP2 expression HUMAN MOLECULAR GENETICS Liu, J., Francke, U. 2006; 15 (11): 1769-1782

    Abstract

    Rett syndrome (RTT) is an X-linked dominant disabling neurodevelopmental disorder caused by loss of function mutations in the MECP2 gene, located at Xq28, which encodes a multifunctional protein. MECP2 expression is regulated in a developmental stage and cell-type-specific manner. The need for tightly controlled MeCP2 levels in brain is strongly suggested by neurologically abnormal phenotypes of mouse models with mild overexpression and by mental retardation in human males with MECP2 duplication. We set out to identify long-range cis-regulatory sequences that differentially regulate MECP2 transcription and, when mutated, may contribute to the pathogenesis of RTT, autism or X-linked mental retardation. By inter-species sequence comparisons, we detected 27 highly conserved non-coding DNA sequences within a 210 kb region covering MECP2. We functionally confirmed four enhancer and two silencer elements by performing luciferase reporter assays in four different human cell lines. The transcription factor binding capability of the identified regulatory elements was tested by gel shift assays. To locate the human MECP2 core promoter, we dissected the promoter region by reporter assays with deletion constructs. We then used chromosome conformation capture methods to document long-range interactions of three enhancers and two silencers with the MECP2 promoter. Acting over distances of up to 130 kb, these elements may influence chromatin configurations and regulate MECP2 transcription. Our study has defined the "MECP2 functional expression module" and identified enhancer and silencer elements that are likely to be responsible for the tissue-specific, developmental stage-specific or splice-variant-specific control of MeCP2 protein expression.

    View details for DOI 10.1093/hmg/ddl099

    View details for Web of Science ID 000237696700003

    View details for PubMedID 16613900

  • Mechanisms of disease: neurogenetics of MeCP2 deficiency NATURE CLINICAL PRACTICE NEUROLOGY Francke, U. 2006; 2 (4): 212-221

    Abstract

    Rett syndrome (RTT) is unique among genetic, chromosomal and other developmental disorders because of its extreme female gender bias, early normal development, and subsequent developmental regression with loss of motor and language skills. RTT is caused by heterozygosity for mutations in the X-linked gene MECP2, which encodes methyl-CpG binding protein 2. MeCP2 is a multifunctional protein that can act as an architectural chromatin-binding protein, a function that is unrelated to its ability to bind methyl-CpG and to attract chromatin modification complexes. Inactivating mutations that cause RTT in females are not prenatally lethal in males, but lead to profound congenital encephalopathy. Molecular diagnoses of RTT, through demonstration of a MECP2 mutation, made at an early stage of the disorder, usually confirm the sporadic nature and very low recurrence risk of the condition. A positive DNA test result, however, also predicts the inevitable clinical course, given the lack of effective intervention. Initial hypotheses indicating that the MeCP2 protein acts as a genome-wide transcriptional repressor were not confirmed by global gene expression studies in various tissues of individuals with RTT and mouse models of MeCP2 deficiency. Rather, recent evidence points to low-magnitude effects of a small number of genes--including the brain--derived neurotrophic factor pathway and glucocorticoid response genes-that might affect formation and maturation of synapses or synaptic function in postmitotic neurons.

    View details for DOI 10.1038/ncpneuro0148

    View details for Web of Science ID 000236359400009

    View details for PubMedID 16932552

  • Inactivating mutations in ESCO2 cause SC phocomelia and Roberts syndrome: No phenotype-genotype correlation AMERICAN JOURNAL OF HUMAN GENETICS Schule, B., Oviedo, A., Johnston, K., Pai, S., Francke, U. 2005; 77 (6): 1117-1128

    Abstract

    The rare, autosomal recessive Roberts syndrome (RBS) is characterized by tetraphocomelia, profound growth deficiency of prenatal onset, craniofacial anomalies, microcephaly, and mental deficiency. SC phocomelia (SC) has a milder phenotype, with a lesser degree of limb reduction and with survival to adulthood. Since heterochromatin repulsion (HR) is characteristic for both disorders and is not complemented in somatic-cell hybrids, it has been hypothesized that the disorders are allelic. Recently, mutations in ESCO2 (establishment of cohesion 1 homolog 2) on 8p21.1 have been reported in RBS. To determine whether ESCO2 mutations are also responsible for SC, we studied three families with SC and two families in which variable degrees of limb and craniofacial abnormalities, detected by fetal ultrasound, led to pregnancy terminations. All cases were positive for HR. We identified seven novel mutations in exons 3-8 of ESCO2. In two families, affected individuals were homozygous--for a 5-nucleotide deletion in one family and a splice-site mutation in the other. In three nonconsanguineous families, probands were compound heterozygous for a single-nucleotide insertion or deletion, a nonsense mutation, or a splice-site mutation. Abnormal splice products were characterized at the RNA level. Since only protein-truncating mutations were identified, regardless of clinical severity, we conclude that genotype does not predict phenotype. Having established that RBS and SC are caused by mutations in the same gene, we delineated the clinical phenotype of the tetraphocomelia spectrum that is associated with HR and ESCO2 mutations and differentiated it from other types of phocomelia that are negative for HR.

    View details for Web of Science ID 000233241200020

    View details for PubMedID 16380922

  • Lack of Pwcr1/MBII-85 snoRNA is critical for neonatal lethality in Prader-Willi syndrome mouse models MAMMALIAN GENOME Ding, F., Prints, Y., Dhar, M. S., Johnson, D. K., Garnacho-Montero, C., Nicholls, R. D., Francke, U. 2005; 16 (6): 424-431

    Abstract

    Prader-Willi syndrome (PWS) is a neurobehavioral disorder caused by the lack of paternal expression of imprinted genes in the human chromosome region 15q11-13. Recent studies of rare human translocation patients narrowed the PWS critical genes to a 121-kb region containing PWCR1/HBII-85 and HBII-438 snoRNA genes. The existing mouse models of PWS that lack the expression of multiple genes, including Snrpn, Ube3a, and many intronic snoRNA genes, are characterized by 80%-100% neonatal lethality. To define the candidate region for PWS-like phenotypes in mice, we analyzed the expression of several genetic elements in mice carrying the large radiation-induced p(30PUb) deletion that includes the p locus. Mice having inherited this deletion from either parent develop normally into adulthood. By Northern blot and RT-PCR assays of brain tissue, we found that Pwcr1/MBII-85 snoRNAs are expressed normally, while MBII-52 snoRNAs are not expressed when the deletion is paternally inherited. Mapping of the distal deletion breakpoint indicated that the p30PUb deletion includes the entire MBII-52 snoRNA gene cluster and three previously unmapped EST sequences. The lack of expression of these elements in mice with a paternal p30PUb deletion indicates that they are not critical for the neonatal lethality observed in PWS mouse models. In addition, we identified MBII-436, the mouse homolog of the HBII-436 snoRNA, confirmed its imprinting status, and mapped it outside of the p30PUb deletion. Taking together all available data, we conclude that the lack of Pwcr1/MBII-85 snoRNA expression is the most likely cause for the neonatal lethality in PWS model mice.

    View details for DOI 10.1007/s00335-005-2460-2

    View details for Web of Science ID 000230639900004

    View details for PubMedID 16075369

  • Molecular breakpoint cloning and gene expression studies of a novel translocation t(4;15)(q27;q11.2) associated with Prader-Willi syndrome BMC MEDICAL GENETICS Schule, B., Albalwi, M., Northrop, E., Francis, D. I., Rowell, M., Slater, H. R., Gardner, R. J., Francke, U. 2005; 6

    Abstract

    Prader-Willi syndrome (MIM #176270; PWS) is caused by lack of the paternally-derived copies, or their expression, of multiple genes in a 4 Mb region on chromosome 15q11.2. Known mechanisms include large deletions, maternal uniparental disomy or mutations involving the imprinting center. De novo balanced reciprocal translocations in 5 reported individuals had breakpoints clustering in SNRPN intron 2 or exon 20/intron 20. To further dissect the PWS phenotype and define the minimal critical region for PWS features, we have studied a 22 year old male with a milder PWS phenotype and a de novo translocation t(4;15)(q27;q11.2).We used metaphase FISH to narrow the breakpoint region and molecular analyses to map the breakpoints on both chromosomes at the nucleotide level. The expression of genes on chromosome 15 on both sides of the breakpoint was determined by RT-PCR analyses.Pertinent clinical features include neonatal hypotonia with feeding difficulties, hypogonadism, short stature, late-onset obesity, learning difficulties, abnormal social behavior and marked tolerance to pain, as well as sticky saliva and narcolepsy. Relative macrocephaly and facial features are not typical for PWS. The translocation breakpoints were identified within SNRPN intron 17 and intron 10 of a spliced non-coding transcript in band 4q27. LINE and SINE sequences at the exchange points may have contributed to the translocation event. By RT-PCR of lymphoblasts and fibroblasts, we find that upstream SNURF/SNRPN exons and snoRNAs HBII-437 and HBII-13 are expressed, but the downstream snoRNAs PWCR1/HBII-85 and HBII-438A/B snoRNAs are not.As part of the PWCR1/HBII-85 snoRNA cluster is highly conserved between human and mice, while no copy of HBII-438 has been found in mouse, we conclude that PWCR1/HBII-85 snoRNAs is likely to play a major role in the PWS- phenotype.

    View details for DOI 10.1186/1471-2350-6-18

    View details for Web of Science ID 000230403600001

    View details for PubMedID 15877813

    View details for PubMedCentralID PMC1142316

  • Frizzled 9 knock-out mice have abnormal B-cell development BLOOD Ranheim, E. A., Kwan, H. C., Reya, T., Wang, Y. K., Weissman, I. L., FRANCKE, U. 2005; 105 (6): 2487-2494

    Abstract

    The binding of frizzled (Fzd) receptors by their Wnt ligands results in the inhibition of beta-catenin degradation and subsequent transcription of beta-catenin/LEF-inducible genes. The beta-catenin pathway is known to be involved in development, tumorigenesis, and stem cell self-renewal. In humans, the FZD9 gene lies in the region of chromosome 7q11.23 deleted in the neurodevelopmental disorder, Williams-Beuren syndrome (WBS). Fzd9-/- mice show no obvious features of WBS, but reveal a role for Fzd9 in lymphoid development and maturation. Fzd9-/- mice show pronounced splenomegaly, thymic atrophy, and lymphadenopathy with age, with accumulation of plasma cells in lymph nodes. There is a depletion of developing B cells in the bone marrow (BM), particularly in the pre-B stage where immunoglobulin heavy chains are expressed and the cells are undergoing clonal expansion prior to light chain rearrangement. The pre-B defect is partially intrinsic to the hematopoietic system; as in competitive BM reconstitution studies, Fzd9-/- -derived BM exhibits defective B-cell development when implanted into a wild-type host. Mature B cells are present in normal numbers in lymph node and spleen. These findings suggest a role for Fzd9 signaling in lymphoid development, particularly at points where B cells undergo self-renewal prior to further differentiation.

    View details for DOI 10.1182/blood-2004-06-2334

    View details for Web of Science ID 000227630500047

    View details for PubMedID 15572594

  • Normal histone modifications on the inactive X chromosome in ICF and Rett syndrome cells: implications for methyl-CpG binding proteins. BMC biology Gartler, S. M., Varadarajan, K. R., Luo, P., Canfield, T. K., Traynor, J., Francke, U., Hansen, R. S. 2004; 2: 21-?

    Abstract

    In mammals, there is evidence suggesting that methyl-CpG binding proteins may play a significant role in histone modification through their association with modification complexes that can deacetylate and/or methylate nucleosomes in the proximity of methylated DNA. We examined this idea for the X chromosome by studying histone modifications on the X chromosome in normal cells and in cells from patients with ICF syndrome (Immune deficiency, Centromeric region instability, and Facial anomalies syndrome). In normal cells the inactive X has characteristic silencing type histone modification patterns and the CpG islands of genes subject to X inactivation are hypermethylated. In ICF cells, however, genes subject to X inactivation are hypomethylated on the inactive X due to mutations in the DNA methyltransferase (DNMT3B) genes. Therefore, if DNA methylation is upstream of histone modification, the histones on the inactive X in ICF cells should not be modified to a silent form. In addition, we determined whether a specific methyl-CpG binding protein, MeCP2, is necessary for the inactive X histone modification pattern by studying Rett syndrome cells which are deficient in MeCP2 function.We show here that the inactive X in ICF cells, which appears to be hypomethylated at all CpG islands, exhibits normal histone modification patterns. In addition, in Rett cells with no functional MeCP2 methyl-CpG binding protein, the inactive X also exhibits normal histone modification patterns.These data suggest that DNA methylation and the associated methyl-DNA binding proteins may not play a critical role in determining histone modification patterns on the mammalian inactive X chromosome at the sites analyzed.

    View details for PubMedID 15377381

  • Normal histone modifications on the inactive X chromosome in ICF and Rett syndrome cells: implications for methyl-CpG binding proteins BMC BIOLOGY Gartler, S. M., Varadarajan, K. R., Luo, P., Canfield, T. K., Traynor, J., Francke, U., Hansen, R. S. 2004; 2
  • Absence of frizzled 9 affects self-renewal signals in developing murine B cells and alters mature B cell and plasma cell function. 45th Annual Meeting and Exhibition of the American-Society-of-Hematology Ranheim, E. A., Kwan, H. C., Wang, Y. K., Reya, T., FRANCKE, U., Weissman, I. L. AMER SOC HEMATOLOGY. 2003: 49A–49A
  • Frizzled 9 knockout mice have defects in B cell development and function 90th Annual Meeting of the American-Association-for-Immunologists Ranheim, E. A., Kwan, H. C., Wang, Y. K., Reya, T., Weissman, I. L., FRANCKE, U. FEDERATION AMER SOC EXP BIOL. 2003: C95–C96
  • Gene expression patterns vary in clonal cell cultures from Rett syndrome females with eight different MECP2 mutations. BMC medical genetics Traynor, J., Agarwal, P., Lazzeroni, L., Francke, U. 2002; 3: 12-?

    Abstract

    Females with the neurological disorder Rett syndrome are heterozygous for mutations in X-linked MECP2 that encodes methyl-CpG binding protein 2 (MeCP2) thought to act as a transcriptional repressor. To identify target genes for MeCP2 modulation, we studied global gene expression in single cell-derived wild-type and mutant MECP2 expressing fibroblast clones with four common mutations (R106W, R306C, 705delG, 1155del32) and in lymphoblastoid cell lines (LCLs) that included four mutant MeCP2 (T158M, 803delG, R168X and 1159del28) expressing, and five (1159del28, R106W, R255X, 803delG, 803delG) wild-type MeCP2 expressing lines.Clonality and mutation status were verified by androgen receptor methylation assays for X-inactivation and by sequencing MECP2 transcripts. Expression studies were done with oligonucleotide microarrays (Affymetrix U95) and verified with real-time quantitative RT-PCR using Sybr Green.Expression of 49 transcripts was increased, and expression of 21 transcripts was decreased, in at least 3 of 4 mutant/wild-type fibroblast comparisons. Transcript levels of 11 genes, determined by quantitative RT-PCR, were highly correlated with the microarray data. Therefore, multiple additional clones from two Rett individuals were tested by RT-PCR only. Striking expression differences were found in both mutant and wildtype MeCP2 expressing clones. Comparing expression profiles of lymphoblastoid cell lines yielded 16 differentially expressed genes.MeCP2 deficiency does not lead to global deregulation of gene expression. Either MeCP2's in vivo function does not involve widespread transcriptional repression, or its function is redundant in cell types that also express other methyl-CpG binding proteins. Our data suggest that clonal fibroblast strains may show substantial inter-strain variation, making them a difficult and unstable resource for genome-wide expression profiling studies.

    View details for PubMedID 12418965

  • Evidence for the role of PWCR1/HBII-85 C/D box small nucleolar RNAs in Prader-Willi syndrome AMERICAN JOURNAL OF HUMAN GENETICS Gallagher, R. C., Pils, B., Albalwi, M., Francke, U. 2002; 71 (3): 669-678

    Abstract

    Prior work has suggested that loss of expression of one or more of the many C/D box small nucleolar RNAs (snoRNAs) encoded within the complex, paternally expressed SNRPN (small nuclear ribonuclear protein N) locus may result in the phenotype of Prader-Willi syndrome (PWS). We suggest that the minimal critical region for PWS is approximately 121 kb within the >460-kb SNRPN locus, bordered by a breakpoint cluster region identified in three individuals with PWS who have balanced reciprocal translocations and by the proximal deletion breakpoint of a familial deletion found in an unaffected mother, her three children with Angelman syndrome, and her father. The subset of SNRPN-encoded snoRNAs within this region comprises the PWCR1/HBII-85 cluster of snoRNAs and the single HBII-438A snoRNA. These are the only known genes within this region, which suggests that loss of their expression may be responsible for much or all of the phenotype of PWS. This hypothesis is challenged by findings in two individuals with PWS who have balanced translocations with breakpoints upstream of the proposed minimal critical region but whose cells were reported to express transcripts within it, adjacent to these snoRNAs. By use of real-time quantitative reverse-transcriptase polymerase chain reaction, we reassessed expression of these transcripts and of the snoRNAs themselves in fibroblasts of one of these patients. We find that the transcripts reported to be expressed in lymphoblast-somatic cell hybrids are not expressed in fibroblasts, and we suggest that the original results were misinterpreted. Most important, we show that the PWCR1/HBII-85 snoRNAs are not expressed in fibroblasts of this individual. These results are consistent with the hypothesis that loss of expression of the snoRNAs in the proposed minimal critical region confers much or all of the phenotype of PWS.

    View details for Web of Science ID 000177489600023

    View details for PubMedID 12154412

  • Premature termination mutations in FBN1: Distinct effects on differential allelic expression and on protein and clinical phenotypes AMERICAN JOURNAL OF HUMAN GENETICS Schrijver, I., Liu, W. G., Odom, R., Brenn, T., Oefner, P., FURTHMAYR, H., Francke, U. 2002; 71 (2): 223-237

    Abstract

    Marfan syndrome (MFS) and other type 1 fibrillinopathies result from mutations in the FBN1 gene, which encodes the connective-tissue microfibrillar protein fibrillin 1. Attempts at correlating genotype with phenotype have suggested considerable heterogeneity. To define the subtype of fibrillinopathy caused by premature termination codon (PTC) mutations, we integrate genotype information and mRNA expression levels with clinical and biochemical phenotypes. By screening the entire FBN1 gene for mutations, we identified 34 probands with PTC mutations. With the exception of two recurrent mutations, these nonsense and frameshift mutations are unique and span the entire FBN1 gene, from IVS2 to IVS63. Allele-specific reverse-transcriptase polymerase chain reaction analyses revealed differential allelic expression in all studied samples, with variable reduction of the mutant transcript. Fibrillin protein synthesis and deposition into the extracellular matrix were studied by pulse-chase analysis of cultured fibroblasts. In the majority of PTC samples, synthesis of normal-sized fibrillin protein was approximately 50% of control levels, but matrix deposition was disproportionately decreased. Probands and mutation-positive relatives were clinically evaluated by means of a standardized protocol. Only 71% (22/31) of probands and 58% (14/24) of the mutation-positive family members met current clinical diagnostic criteria for MFS. When compared with our previously reported study group of 44 individuals with FBN1 cysteine substitutions, the PTC group showed statistically significant differences in the frequency of individual signs, especially in the ocular manifestations. Whereas large-joint hypermobility was more common, lens dislocation and retinal detachment were distinctly less common in the PTC group. We conclude that PTC mutations have a major impact on the pathogenesis of type 1 fibrillinopathies and convey a distinct biochemical, clinical, and prognostic profile.

    View details for Web of Science ID 000176977700002

    View details for PubMedID 12068374

  • Spontaneous spinal cerebrospinal fluid leaks and minor skeletal features of Marfan syndrome: a microfibrillopathy JOURNAL OF NEUROSURGERY Schrijver, I., Schievink, W. I., Godfrey, M., Meyer, F. B., FRANCKE, U. 2002; 96 (3): 483-489

    Abstract

    Spontaneous spinal cerebrospinal fluid (CSF) leaks are increasingly recognized as a cause of postural headaches. The authors examined a group of patients suffering from spontaneous spinal CSF leaks who also had minor skeletal features of Marfan syndrome for abnormalities of fibrillin-containing microfibrils.Patients with spontaneous CSF leaks were evaluated for the clinical characteristics of connective tissue disorders. Skin biopsies were obtained in three patients with skeletal manifestations that constitute part of the Marfan syndrome phenotype. Cultured fibroblasts were studied for fibrillin-1 synthesis and incorporation into the extracellular matrix (ECM) by performing quantitative metabolic labeling and immunohistochemical analysis. Among 20 consecutive patients found to have spinal CSF leaks, four (20%) exhibited minor skeletal features of Marfan syndrome, but lacked any ocular or cardiovascular abnormalities. The mean age of these patients (30 years) was lower than that of the 16 patients without skeletal abnormalities (44 years; p = 0.01). Abnormalities in fibrillin-1 metabolism and immunostaining were detected in all three patients with the skeletal abnormalities who underwent examination, but not in a control patient without these skeletal manifestations.Twenty percent of patients who experience spontaneous spinal CSF leaks have minor skeletal features of Marfan syndrome. The authors demonstrated abnormalities in fibrillin-1 protein deposition in all patients examined, but only one person was found to have a fibrillin-1 abnormality typically found in classic Marfan syndrome. The results indicate that there is a heterogeneous involvement of other components of ECM microfibrils at the basis of this cerebrospinal manifestation. In addition, the authors identified a connective-tissue etiological factor in a group of disorders not previously classified as such.

    View details for Web of Science ID 000174104100004

    View details for PubMedID 11883832

  • Spectrum of MECP2 mutations in Rett syndrome World Congress on Rett Syndrome Lee, S. S., Wan, M. M., Francke, U. ELSEVIER SCIENCE BV. 2001: S138–S143

    Abstract

    Mutations in the methyl-CpG-binding protein 2 gene (MECP2) are identified in the majority of females with Rett syndrome (RTT), an X-linked dominant neurodevelopmental disorder. We searched for mutations by sequencing the MECP2 coding region in 45 sporadic cases (35 with classic RTT, eight with variant forms and two males) and in seven families with two or more affected females. Following our previous report of mutations in two families and eight sporadic cases, we here present 18 additional mutations. We found 13 single nucleotide substitutions, all of which are C-->T transitions at CpG hot spots. Frameshift mutations, leading to premature termination of translation, include two single guanine (G) nucleotide deletions from a stretch of contiguous Gs, a novel four nucleotide deletion, a novel 32 nucleotide deletion in the C-terminal domain and a novel complex duplication/deletion rearrangement in the same region. When X-chromosome inactivation patterns were compared in 16 MECP2 mutation-positive and 23 mutation-negative samples, no significant differences were observed. The mutational spectrum in our subject population is similar to studies from around the world. Of over 300 MECP2 mutations reported, two-thirds are truncating mutations and one-third are missense mutations, mostly in the methyl-binding domain. Nearly 70% of all identified mutations are C-->T transitions at one of eight CpG hot spots, and about 10% are intragenic deletions or complex rearrangements that lead to frameshifts in the C-terminal region. The rate of mutation detection in the MECP2 coding region ranges from 70 to 85% in clinically diagnosed RTT and is much lower in diagnostic variants.

    View details for Web of Science ID 000173341900028

    View details for PubMedID 11738860

  • frizzled 9 is expressed in neural precursor cells in the developing neural tube DEVELOPMENT GENES AND EVOLUTION Van Raay, T. J., Wang, Y. K., Stark, M. R., Rasmussen, J. T., FRANCKE, U., Vetter, M. L., Rao, M. S. 2001; 211 (8-9): 453-457

    Abstract

    The wnt signaling pathway has important functions in nervous system development. To better understand this process we have cloned and analyzed the expression of the wnt receptor, frizzled 9, in the developing nervous system in mouse, chick and zebrafish. The earliest expression of mouse frizzled 9 mRNA expression begins at E8.5 with expression throughout the entire rostral-caudal neuraxis. This early expression pattern within the neural tube appears to be conserved between chick and zebrafish. Expression becomes restricted to a ventral domain in the mouse ventricular zone at E11.5, a region specified to give rise to neurons and glia. Using a polyclonal antibody to MFZ9 further shows expression limited to neural restricted precursors cells.

    View details for Web of Science ID 000171330300013

    View details for PubMedID 11685582

  • Guidelines for reporting clinical features in cases with MECP2 mutations BRAIN & DEVELOPMENT Kerr, A. M., Nomura, Y., Armstrong, D., ANVRET, M., Belichenko, P. V., Budden, S., Cass, H., Christodoulou, J., Clarke, A., Ellaway, C., D'Esposito, M., FRANCKE, U., Hulten, M., Julu, P., Leonard, H., Naidu, S., Schanen, C., Webb, T., Engerstrom, I. W., Yamashita, Y., Segawa, M. 2001; 23 (4): 208-211

    Abstract

    An international group recommends that papers relating phenotypes to genotypes involving mutations in the X chromosome gene MECP2 should provide a minimum data set reporting the range of disturbances frequently encountered in Rett Syndrome. A simple scoring system is suggested which will facilitate comparison among the various clinical profiles. Features are described which should prompt screening for MECP2 mutations.

    View details for Web of Science ID 000169249300002

    View details for PubMedID 11376997

  • Evolutionary relationships among Rel domains indicate functional diversification by recombination PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Graef, I. A., Gastier, J. M., FRANCKE, U., Crabtree, G. R. 2001; 98 (10): 5740-5745

    Abstract

    The recent sequencing of several complete genomes has made it possible to track the evolution of large gene families by their genomic structure. Following the large-scale association of exons encoding domains with well defined functions in invertebrates could be useful in predicting the function of complex multidomain proteins in mammals produced by accretion of domains. With this objective, we have determined the genomic structure of the 14 genes in invertebrates and vertebrates that contain rel domains. The sequence encoding the rel domain is defined by intronic boundaries and has been recombined with at least three structurally and functionally distinct genomic sequences to generate coding sequences for: (i) the rel/Dorsal/NFkappaB proteins that are retained in the cytoplasm by IkB-like proteins; (ii) the NFATc proteins that sense calcium signals and undergo cytoplasmic-to-nuclear translocation in response to dephosphorylation by calcineurin; and (iii) the TonEBP tonicity-responsive proteins. Remarkably, a single exon in each NFATc family member encodes the entire Ca(2+)/calcineurin sensing region, including nuclear import/export, calcineurin-binding, and substrate regions. The Rel/Dorsal proteins and the TonEBP proteins are present in Drosophila but not Caenorhabditis elegans. On the other hand, the calcium-responsive NFATc proteins are present only in vertebrates, suggesting that the NFATc family is dedicated to functions specific to vertebrates such as a recombinational immune response, cardiovascular development, and vertebrate-specific aspects of the development and function of the nervous system.

    View details for Web of Science ID 000168623300067

    View details for PubMedID 11344309

  • MECP2 truncating mutations cause histone H4 hyperacetylation in Rett syndrome HUMAN MOLECULAR GENETICS Wan, M. M., Zhao, K. J., Lee, S. S., FRANCKE, U. 2001; 10 (10): 1085-1092

    Abstract

    Rett syndrome (RTT) is a mostly sporadic disorder of developmental regression, with loss of speech and purposeful hand use, microcephaly and seizures. It affects 1 in 10 000-15 000 females. RTT is caused by mutations in the MECP2 gene, which is located in Xq28 and subject to X inactivation. MECP2 encodes a methyl-CpG-binding protein that binds to 5-methyl-cytosine in DNA through its methyl-binding domain. Recruitment of a transcriptional silencing complex through MeCP2's transcriptional repression domain results in histone deacetylation and chromatin condensation. To study the effects of two common truncating RTT mutations (R168X and 803delG), we examined mutant MeCP2 expression and global histone acetylation levels in clonal cell cultures from a female RTT patient with the mutant R168X allele on the active X chromosome, as well as in cells from a male hemizygous for the frameshift mutation 803delG (V288X). Both mutant alleles generated stable RNA transcripts, but no intact MeCP2 protein was detected with an antibody against the C-terminal region of MeCP2. Western blots with antibodies against acetylated histones H3 and H4 revealed that H4, but not H3, was hyperacetylated. By using antibodies against individual acetylated lysine residues, the observed H4 hyperacetylation was attributed to increased acetylation of lysine 16. Therefore, expression of endogenous truncating MECP2 alleles, in the absence of wild-type MeCP2 protein, is specifically associated with an increase in the mono-acetylated histone isoform H4K16. This observed effect may result in over-expression of MeCP2 target genes and, thus, play a role in the pathogenesis of RTT.

    View details for Web of Science ID 000168773700009

    View details for PubMedID 11331619

  • Human genetics around the world GENETICS IN MEDICINE FRANCKE, U. 2001; 3 (3): 167-167

    View details for Web of Science ID 000171007700002

    View details for PubMedID 11388755

  • Association of acetylated histones with paternally expressed genes in the Prader-Willi deletion region HUMAN MOLECULAR GENETICS Fulmer-Smentek, S. B., FRANCKE, U. 2001; 10 (6): 645-652

    Abstract

    Imprinted genes within the Prader-Willi/Angelman syndrome region of human chromosome 15q11-q13 are regulated by a mechanism involving allele-specific DNA methylation. Since transcriptional regulation by DNA methylation involves histone deacetylation, we explored whether differences in histone acetylation exist between the two parental alleles of SNRPN and other paternally expressed genes in the region by using a chromatin immunoprecipitation assay with antibodies against acetylated histones H3 and H4. SNRPN exon 1, which is methylated on the silent maternal allele, was associated with acetylated histones on the expressed paternal allele only. SNRPN intron 7, which is methylated on the paternal allele, was not associated with acetylated histones on either allele. The paternally expressed genes NDN, IPW, PWCR1 and MAGEL2 were not associated with acetylated histones on either allele. Treatment of the lymphoblastoid cells with trichostatin A, a histone deacetylase inhibitor, did not result in any changes to SNRPN expression or association of acetylated histones with exon 1. Treatment with 5-aza-deoxycytidine (5-aza-dC), which inhibits DNA methylation, resulted in activation of SNRPN expression from the maternal allele, but was not accompanied by acetylation of histones. Our finding of allele-specific association of acetylated histones with the SNRPN exon 1 region, which encompasses the imprinting center, suggests that histone acetylation at this site may be important for regulation of SNRPN and of other paternally expressed genes in the region. On the silent allele, 5-aza-dC treatment altered SNRPN expression, but not association with acetylated histones, suggesting that histone acetylation is a secondary event in the process of gene reactivation by CpG demethylation.

    View details for Web of Science ID 000167533800011

    View details for PubMedID 11230184

  • Williams (Williams Beuren) syndrome: A distinct neurobehavioral disorder JOURNAL OF CHILD NEUROLOGY Kaplan, P., Wang, P. P., FRANCKE, U. 2001; 16 (3): 177-190

    View details for Web of Science ID 000171025500005

    View details for PubMedID 11305686

  • The human genome project: implications for the endocrinologist. Journal of pediatric endocrinology & metabolism FRANCKE, U. 2001; 14: 1395-1408

    Abstract

    The sequencing of the human genome is a major achievement of our time. This article reviews the process and current status of the working draft sequence, ways to predict genes and assign function, and conclusions for human biology. Gene density is uneven and related to chromosome banding patterns, and the estimate of approximately 30,000 genes is lower than expected. Genetic maps for men and women differ from each other and from the physical map. Single nucleotide polymorphisms occur at an average spacing of 1 kb. Human populations are 99.99% identical, and most sequences are shared between people from different continents. To illustrate the tools for accessing the human genome sequence, searches were performed for genes encoding three categories of growth-related proteins, insulin-like growth factor-I (IGF-I) receptor, IGF-binding proteins and growth hormone receptor. The results revealed novel details about their genomic organization and new predicted transcripts. Impacts on medicine are promised in the fields of diagnostics (development of new tests), therapeutics (identification of new potential drug targets) and pharmacogenomics (streamlining of drug discovery and personalized medicine). Associated ethical, legal and social implications and controversies include genetic determinism, informed consent, privacy and confidentiality, ownership of genetic information in the biotechnology marketplace, and access to genetic healthcare.

    View details for PubMedID 11837493

  • Multi-exon deletions of the FBN1 gene in Marfan syndrome. BMC medical genetics Liu, W., Schrijver, I., Brenn, T., FURTHMAYR, H., FRANCKE, U. 2001; 2: 11-?

    Abstract

    Mutations in the fibrillin -1 gene (FBN1) cause Marfan syndrome (MFS), an autosomal dominant multi-system connective tissue disorder. The 200 different mutations reported in the 235 kb, 65 exon-containing gene include only one family with a genomic multi-exon deletion.We used long-range RT-PCR for mutation detection and long-range genomic PCR and DNA sequencing for identification of deletion breakpoints, allele-specific transcript analyses to determine stability of the mutant RNA, and pulse-chase studies to quantitate fibrillin synthesis and extracellular matrix deposition in cultured fibroblasts. Southern blots of genomic DNA were probed with three overlapping fragments covering the FBN1 coding exonsTwo novel multi-exon FBN1 deletions were discovered. Identical nucleotide pentamers were found at or near the intronic breakpoints. In a Case with classic MFS, an in-frame deletion of exons 42 and 43 removed the C-terminal 24 amino acids of the 5th LTBP (8-cysteine) domain and the adjacent 25th calcium-binding EGF-like (6-cysteine) domain. The mutant mRNA was stable, but fibrillin synthesis and matrix deposition were significantly reduced. A Case with severe childhood-onset MFS has a de novo deletion of exons 44-46 that removed three EGF-like domains. Fibrillin protein synthesis was normal, but matrix deposition was strikingly reduced. No genomic rearrangements were detected by Southern analysis of 18 unrelated MFS samples negative for FBN1 mutation screening.Two novel deletion cases expand knowledge of mutational mechanisms and genotype/phenotype correlations of fibrillinopathies. Deletions or mutations affecting an LTBP domain may result in unstable mutant protein cleavage products that interfere with microfibril assembly.

    View details for PubMedID 11710961

  • Small evolutionarily conserved RNA, resembling C/D box small nucleolar RNA, is transcribed from PWCR1, a novel imprinted gene in the Prader-Willi deletion region, which is highly expressed in brain AMERICAN JOURNAL OF HUMAN GENETICS de los Santos, T., Schweizer, J., Rees, C. A., Francke, U. 2000; 67 (5): 1067-1082

    Abstract

    Prader-Willi syndrome is a complex neurodevelopmental disorder caused by the inactivation or deletion of imprinted, paternally expressed genes in chromosome band 15q11.2. We report the identification and characterization of PWCR1, a novel imprinted gene within that region, and its mouse orthologue, Pwcr1, which was mapped to the conserved syntenic region on mouse chromosome 7. Expressed only from the paternal allele, both genes require the imprinting-center regulatory element for expression and are transcribed from the same strand. They are intronless and do not appear to encode a protein product. High human/mouse sequence similarity (87% identity) is limited to a 99-bp region called "HMCR" (for "human-mouse conserved region"). The HMCR sequence has features of a C/D box small nucleolar RNA (snoRNA) and is represented in an abundant small transcript in both species. Located in nucleoli, snoRNAs serve as methylation guidance RNAs in the modification of ribosomal RNA and other small nuclear RNAs. In addition to the nonpolyadenylated small RNAs, larger polyadenylated PWCR1 transcripts are found in most human tissues, whereas expression of any Pwcr1 RNAs is limited to mouse brain. Genomic sequence analysis reveals the presence of multiple copies of PWCR1 and Pwcr1 that are organized within local tandem-repeat clusters. On a multispecies Southern blot, hybridization to an HMCR probe encoding the putative snoRNA is limited to mammals.

    View details for Web of Science ID 000165091600004

    View details for PubMedID 11007541

  • Discovery of the Rett syndrome gene and its function TURKISH JOURNAL OF PEDIATRICS FRANCKE, U. 2000; 42 (4): 271-271

    View details for Web of Science ID 000166335400001

    View details for PubMedID 11196739

  • Rett syndrome is caused by mutations in the X-linked MECP2 gene encoding methyl-CpG-binding protein Zoghbi, H. Y., Amir, R. E., Wan, M., Lee, S. S., Van den Veyver, I. B., Tran, C. Q., Malicki, D., Schanen, N. C., FRANCKE, U. CELL PRESS. 2000: 1723–23
  • Manifestations and linkage analysis in X-linked autoimmunity-immunodeficiency syndrome AMERICAN JOURNAL OF MEDICAL GENETICS Ferguson, P. J., Blanton, S. H., Saulsbury, F. T., McDuffie, M. J., Lemahieu, V., Gastier, J. M., FRANCKE, U., Borowitz, S. M., Sutphen, J. L., Kelly, T. E. 2000; 90 (5): 390-397

    Abstract

    The clinical findings of a kindred with an X-linked disorder are characterized by autoimmune polyendocrinopathy, enteropathy with villous atrophy, chronic dermatitis, and variable immunodeficiency. Linkage analysis was performed on 20 members of the affected kindred to determine the location of the responsible locus. Informative recombinations limited the region to an approximate 20 cM interval bordered by DXS1055 and DXS1196/DXS1050. Multipoint analysis generated a lod score >3 for the region contained between DXS8024 and DXS8031. The candidate region includes the Wiskott-Aldrich syndrome (WAS) locus. Evaluation of the Wiskott-Aldrich syndrome protein gene by single strand conformational analysis, heteroduplex analysis, and direct sequencing of the 12 exons in an affected male and two carrier females revealed no abnormalities. We conclude that this kindred has an X-linked disorder, distinct from WAS, that results in autoimmunity and variable immunodeficiency. The responsible locus maps to the pericentromeric region Xp11.23 to Xq21.1.

    View details for Web of Science ID 000085166300009

    View details for PubMedID 10706361

  • Expression of human Wiskott-Aldrich syndrome protein in patients' cells leads to partial correction of a phenotypic abnormality of cell surface glycoproteins GENE THERAPY Huang, M. M., TSUBOI, S., Wong, A., Yu, X. J., Oh-Eda, M., Derry, J. M., FRANCKE, U., Fukuda, M., Weinberg, K. I., Kohn, D. B. 2000; 7 (4): 314-320

    Abstract

    The Wiskott-Aldrich syndrome (WAS) is an uncommon X-linked recessive disease characterized by thrombocytopenia, eczema and immunodeficiency. The biochemical defect of this disorder primarily affects cells derived from bone marrow. To understand better the molecular mechanisms underlying this disease and to evaluate the possibility of correcting the genetic defects in hematopoietic cells, a Moloney murine leukemia virus (MoMLV)- based retroviral vector carrying a functional Wiskott-Aldrich syndrome protein (WASp) cDNA driven by an SV40 promoter (LNS-WASp) was constructed. A packaging cell line containing this vector produced a stable level of WAS protein and maintained a high titer of viral output. Epstein-Barr virus (EBV)-transformed B lymphoblastoid cell lines (B-LCL) from WAS patients, which lack expression of the WAS protein, were transduced by the LNS-WASp retroviral vector and showed expression of WASp by Western blot. Analysis of the O-glycan pattern on cell surface glycoproteins from WAS patients' B-LCL showed an altered glycosylation pattern, due to increased activity of beta-1, 6-N-acetylglucosaminyltransferase (C2GnT). Transduction by the retroviral vector carrying the functional WASp cDNA partially restored the abnormal glycosylation pattern, and was accompanied by a decreasing C2GnT activity. These findings imply a functional linkage between the WAS protein and the expression of the glycosyltransferase involved in the O-glycosylation, and also suggest a potential gene therapy via transferring a functional WASp cDNA into hematopoietic cells for Wiskott-Aldrich syndrome. Gene Therapy (2000) 7, 314-320.

    View details for Web of Science ID 000085379500008

    View details for PubMedID 10694812

  • A physical map, including a BAC/PAC clone contig, of the Williams-Beuren syndrome-deletion region at 7q11.23 AMERICAN JOURNAL OF HUMAN GENETICS Peoples, R., Franke, Y., Wang, Y. K., Perez-Jurado, L., Paperna, T., Cisco, M., FRANCKE, U. 2000; 66 (1): 47-68

    Abstract

    Williams-Beuren syndrome (WBS) is a developmental disorder caused by haploinsufficiency for genes in a 2-cM region of chromosome band 7q11.23. With the exception of vascular stenoses due to deletion of the elastin gene, the various features of WBS have not yet been attributed to specific genes. Although >/=16 genes have been identified within the WBS deletion, completion of a physical map of the region has been difficult because of the large duplicated regions flanking the deletion. We present a physical map of the WBS deletion and flanking regions, based on assembly of a bacterial artificial chromosome/P1-derived artificial chromosome contig, analysis of high-throughput genome-sequence data, and long-range restriction mapping of genomic and cloned DNA by pulsed-field gel electrophoresis. Our map encompasses 3 Mb, including 1.6 Mb within the deletion. Two large duplicons, flanking the deletion, of >/=320 kb contain unique sequence elements from the internal border regions of the deletion, such as sequences from GTF2I (telomeric) and FKBP6 (centromeric). A third copy of this duplicon exists in inverted orientation distal to the telomeric flanking one. These duplicons show stronger sequence conservation with regard to each other than to the presumptive ancestral loci within the common deletion region. Sequence elements originating from beyond 7q11.23 are also present in these duplicons. Although the duplicons are not present in mice, the order of the single-copy genes in the conserved syntenic region of mouse chromosome 5 is inverted relative to the human map. A model is presented for a mechanism of WBS-deletion formation, based on the orientation of duplicons' components relative to each other and to the ancestral elements within the deletion region.

    View details for Web of Science ID 000085033600007

    View details for PubMedID 10631136

  • Diverse deletions in the growth hormone receptor gene cause growth hormone insensitivity syndrome HUMAN MUTATION Gastier, J. M., Berg, M. A., Vesterhus, P., Reiter, E. O., Francke, U. 2000; 16 (4): 323-333

    Abstract

    Growth hormone insensitivity syndrome (GHIS; also known as Laron syndrome), is characterized by severe postnatal growth failure and normal growth hormone. The syndrome is frequently caused by point mutations in the growth hormone receptor gene (GHR). Here we report five families with GHIS and partial deletions of the GHR gene. The deletion breakpoints were sequenced and PCR-based diagnostic tests were developed. In a Cambodian family, a novel deletion removed part of exon 5 and 1.2 kb of the preceding intron. The deletion occurred by recombination within four identical nucleotides. In the mutant transcript, skipping of the truncated exon 5 leads to a frameshift and premature termination codon (PTC). A previously reported discontinuous deletion of GHR exons 3, 5, and 6 was identified in three Oriental Jewish families. An unaffected individual was heterozygous for the exon 5 and 6 deletion, but homozygously deleted for exon 3 suggesting that the exon 3 deletion is a polymorphism. The pathogenic deletion of exons 5 and 6 spans about 7.5 kb. Sequence analysis of the breakpoints revealed an imperfect junction between introns 4 and 6, with a four basepair insertion. A novel deletion of 13 nucleotides within exon 9 was identified in a Caucasian girl with GHIS who carries the I153T missense mutation on her other allele. The exon 9 deletion leads to a frameshift and PTC. The predicted protein retains the transmembrane domain and a short cytoplasmic tail. Four family members in three generations were carriers of this deletion, but only two of them were below normal for height, suggesting that this mutation by itself does not act as a dominant negative, as was reported for two other GHR mutations which lead to truncation of the intracellular domain.

    View details for Web of Science ID 000089746800005

    View details for PubMedID 11013443

  • Growth hormone receptor deficiency in Ecuador. journal of clinical endocrinology and metabolism Rosenbloom, A. L., Guevara-Aguirre, J., Rosenfeld, R. G., FRANCKE, U. 1999; 84 (12): 4436-4443

    View details for PubMedID 10599699

  • Rett syndrome and beyond: Recurrent spontaneous and familial MECP2 mutations at CpG hotspots AMERICAN JOURNAL OF HUMAN GENETICS Wan, M. M., Lee, S. S., Zhang, X. Y., Houwink-Manville, I., Song, H. R., Amir, R. E., Budden, S., Naidu, S., Pereira, J. L., Lo, I. F., Zoghbi, H. Y., Schanen, N. C., Francke, U. 1999; 65 (6): 1520-1529

    Abstract

    Rett syndrome (RTT) is a neurodevelopmental disorder characterized by loss of acquired skills after a period of normal development in infant girls. The responsible gene, encoding methyl-CpG binding protein 2 (MeCP2), was recently discovered. Here we explore the spectrum of phenotypes resulting from MECP2 mutations. Both nonsense (R168X and R255X) and missense (R106W and R306C) mutations have been found, with multiple recurrences. R168X mutations were identified in six unrelated sporadic cases, as well as in two affected sisters and their normal mother. The missense mutations were de novo and affect conserved domains of MeCP2. All of the nucleotide substitutions involve C-->T transitions at CpG hotspots. A single nucleotide deletion, at codon 137, that creates a L138X stop codon within the methyl-binding domain was found in an individual with features of RTT and incontinentia pigmenti. An 806delG deletion causing a V288X stop in the transcription-repression domain was identified in a woman with motor-coordination problems, mild learning disability, and skewed X inactivation; in her sister and daughter, who were affected with classic RTT; and in her hemizygous son, who died from congenital encephalopathy. Thus, some males with RTT-causing MECP2 mutations may survive to birth, and female heterozygotes with favorably skewed X-inactivation patterns may have little or no involvement. Therefore, MECP2 mutations are not limited to RTT and may be implicated in a much broader phenotypic spectrum.

    View details for Web of Science ID 000084282500006

    View details for PubMedID 10577905

  • Rett syndrome is caused by mutations in X-linked MECP2, encoding methyl-CpG-binding protein 2 NATURE GENETICS Amir, R. E., Van den Veyver, I. B., Wan, M., Tran, C. Q., FRANCKE, U., Zoghbi, H. Y. 1999; 23 (2): 185-188

    Abstract

    Rett syndrome (RTT, MIM 312750) is a progressive neurodevelopmental disorder and one of the most common causes of mental retardation in females, with an incidence of 1 in 10,000-15,000 (ref. 2). Patients with classic RTT appear to develop normally until 6-18 months of age, then gradually lose speech and purposeful hand use, and develop microcephaly, seizures, autism, ataxia, intermittent hyperventilation and stereotypic hand movements. After initial regression, the condition stabilizes and patients usually survive into adulthood. As RTT occurs almost exclusively in females, it has been proposed that RTT is caused by an X-linked dominant mutation with lethality in hemizygous males. Previous exclusion mapping studies using RTT families mapped the locus to Xq28 (refs 6,9,10,11). Using a systematic gene screening approach, we have identified mutations in the gene (MECP2 ) encoding X-linked methyl-CpG-binding protein 2 (MeCP2) as the cause of some cases of RTT. MeCP2 selectively binds CpG dinucleotides in the mammalian genome and mediates transcriptional repression through interaction with histone deacetylase and the corepressor SIN3A (refs 12,13). In 5 of 21 sporadic patients, we found 3 de novo missense mutations in the region encoding the highly conserved methyl-binding domain (MBD) as well as a de novo frameshift and a de novo nonsense mutation, both of which disrupt the transcription repression domain (TRD). In two affected half-sisters of a RTT family, we found segregation of an additional missense mutation not detected in their obligate carrier mother. This suggests that the mother is a germline mosaic for this mutation. Our study reports the first disease-causing mutations in RTT and points to abnormal epigenetic regulation as the mechanism underlying the pathogenesis of RTT.

    View details for Web of Science ID 000082827500017

    View details for PubMedID 10508514

  • Cysteine substitutions in epidermal growth factor-like domains of fibrillin-1: Distinct effects on biochemical and clinical phenotypes AMERICAN JOURNAL OF HUMAN GENETICS Schrijver, I., Liu, W., Brenn, T., FURTHMAYR, H., Francke, U. 1999; 65 (4): 1007-1020

    Abstract

    Fibrillin-1 (FBN1) contains 47 epidermal growth factor (EGF)-like domains characterized by six conserved cysteine residues. Cysteine substitutions that disrupt one of the three disulfide bonds are frequent causes of Marfan syndrome (MFS). We identified 19 new substitutions involving cysteine residues in each of the six positions of EGF-like domains. Allele-specific mRNA assays revealed equal abundance of mutant and normal FBN1 transcripts in all 10 individuals studied. Quantitative pulse-chase analysis of fibrillin protein was performed on 25 mutant fibroblast strains with substitutions of 22 different cysteine residues in 18 different EGF-like domains spanning the entire gene. Normal synthesis and stability of mutant fibrillin molecules was seen in 20/25 individuals, 11 of whom showed delayed intracellular processing and/or secretion. In the remaining five cases, the mutant protein was apparently unstable. In four of these five cases, the second or third disulfide bond of EGF-like domains immediately preceding an 8-cysteine or hybrid domain was affected. All but two mutations caused severe reduction of matrix deposition, which was attributed to a dominant-negative effect of mutant molecules. For genotype/phenotype comparisons, clinical data on 25 probands and 19 mutation-positive family members were analyzed. Ocular manifestations were among the most consistent features (ectopia lentis in 86%, myopia in 80%). Nine mutations encoded by exons 26-32 resulted in early-onset classic MFS and, in one case, neonatal-lethal MFS. Mutations outside this region were associated with variable clinical phenotypes, including individuals with fibrillinopathies not meeting diagnostic criteria for MFS.

    View details for Web of Science ID 000082910000009

    View details for PubMedID 10486319

  • Characterization and expression pattern of the frizzled gene Fzd9, the mouse homolog of FZD9 which is deleted in Williams-Beuren syndrome GENOMICS Wang, Y. K., Sporle, R., Paperna, T., Schughart, K., Francke, U. 1999; 57 (2): 235-248

    Abstract

    The frizzled gene family is conserved from insects to mammals and codes for putative Wnt receptors that share a cysteine-rich extracellular domain and seven transmembrane domains. We previously identified a novel frizzled gene, FZD3, now renamed FZD9, in the Williams-Beuren syndrome (WBS) deletion region at chromosomal band 7q11.23 and showed that its product can interact with the Drosophila wingless protein. Here, we report the characterization of the mouse homolog Fzd9. The Fzd9 gene produces a 2.4-kb transcript encoding a 592-amino-acid protein with 95% identity to the human FZD9. Fzd9 was mapped to the conserved syntenic region on distal mouse chromosome 5. By RNA in situ hybridization studies of whole-mount embryos and sections we delineated the temporal and spatial expression patterns in the neural tube, trunk skeletal muscle precursors (myotomes), limb skeletal anlagen, craniofacial regions, and nephric ducts. In adult mouse tissue, the Fzd9 transcript is abundantly present in heart, brain, testis, and skeletal muscle. In testis, Fzd9 is expressed in all spermatogenic cell types. Immunohistochemical studies of cells transfected with a Fzd9 expression construct confirm that Fzd9 is a membrane protein. These results suggest potential Wnt ligands of Fzd9, a role of Fzd9 in skeletal muscle specification, and contributions of FZD9 to the WBS phenotype.

    View details for Web of Science ID 000079868900006

    View details for PubMedID 10198163

  • The human neuregulin-2 (NRG2) gene: cloning, mapping and evaluation as a candidate for the autosomal recessive form of Charcot-Marie Tooth disease linked to 5q HUMAN GENETICS Ring, H. Z., Chang, H., Guilbot, A., Brice, A., LeGuern, E., FRANCKE, U. 1999; 104 (4): 326-332

    Abstract

    Neuregulin-2 (NRG2) is a novel member of the neuregulin family of growth and differentiation factors. Through interaction with the ErbB family of receptors, neuregulin-2 induces the growth and differentiation of epithelial, neuronal, glial and other types of cells. In this study, we have cloned the human neuregulin-2 gene, and determined its genomic structure and alternative splicing patterns. By using radiation hybrid mapping panels, the human NRG2 gene was mapped to the D5S658-D5S402 region within 5q23-q33, close to an autosomal recessive form of demyelinating Charcot-Marie-Tooth (CMT) disease. The NRG2 gene was found to be on two yeast artificial chromosomes overlapping the candidate interval and was, thus, considered a good positional candidate for this form of CMT. When the entire neuregulin-2 coding sequence and splice junctions were explored, however, no mutation was identified in one CMT family linked to 5q23-q33. In addition, three intronic single nucleotide polymorphisms were identified in the NRG2 gene. Genotyping in two families localized the NRG2 gene outside of the revised candidate interval between D5S402-D5S210 and excluded NRG2 as the gene responsible for this form of CMT disease.

    View details for PubMedID 10369162

  • In vivo nuclease hypersensitivity studies reveal multiple sites of parental origin-dependent differential chromatin conformation in the 150 kb SNRPN transcription unit HUMAN MOLECULAR GENETICS Schweizer, J., Zynger, D., Francke, U. 1999; 8 (4): 555-566

    Abstract

    Human chromosome region 15q11-q13 contains a cluster of oppositely imprinted genes. Loss of the paternal or the maternal alleles by deletion of the region or by uniparental disomy 15 results in Prader-Willi syndrome (PWS) or Angelman syndrome (AS), respectively. Hence, the two phenotypically distinct neurodevelopmental disorders are caused by the lack of products of imprinted genes. Subsets of PWS and AS patients exhibit 'imprinting mutations', such as small microdeletions within the 5' region of the small nuclear ribonucleoprotein polypeptide N ( SNRPN ) transcription unit which affect the transcriptional activity and methylation status of distant imprinted genes throughout 15q11-q13 in cis. To elucidate the mechanism of these long-range effects, we have analyzed the chromatin structure of the 150 kb SNRPN transcription unit for DNase I- and Msp I-hypersensitive sites. By using an in vivo approach on lymphoblastoid cell lines from PWS and AS individuals, we discovered that the SNRPN exon 1 is flanked by prominent hypersensitive sites on the paternal allele, but is completely inaccessible to nucleases on the maternal allele. In contrast, we identified several regions of increased nuclease hypersensitivity on the maternal allele, one of which coincides with the AS minimal microdeletion region and another lies in intron 1 immediately downstream of the paternal-specific hypersensitive sites. At several sites, parental origin-specific nuclease hypersensitivity was found to be correlated with hypermethylation on the allele contributed by the other parent. The differential parental origin-dependent chromatin conformations might govern access of regulatory protein complexes and/or RNAs which could mediate interaction of the region with other genes.

    View details for Web of Science ID 000079477000001

    View details for PubMedID 10072422

  • Mapping of the KHSRP gene to a region of conserved synteny on human chromosome 19p13.3 and mouse chromosome 17 GENOMICS Ring, H. Z., Vameghi-Meyers, V., Nikolic, J. M., Min, H. S., Black, D. L., FRANCKE, U. 1999; 56 (3): 350-352

    Abstract

    The K homology-type splicing regulatory protein, KSRP, activates splicing through intronic splicing enhancer sequences. It is highly expressed in neural cells and is required for the neural-specific splicing of the c-src N1 exon. In this study, we mapped the gene (gene symbols KHSRP and Khsrp) to human chromosome 19 by using radiation hybrid panels and to mouse chromosome 17 by studying an interspecific backcross panel. Human KHSRP is a positional candidate gene for familial febrile convulsion and Cayman type cerebellar ataxia. Comparative analysis of the human and mouse genomes indicates that the KHSRP gene is located in regions of conserved synteny between the two species.

    View details for Web of Science ID 000079390000015

    View details for PubMedID 10087204

  • The mouse Fubp gene maps near the distal end of chromosome 3 GENOMICS Ring, H. Z., Vameghi-Meyers, V., Min, H. S., Nikolic, J. M., Black, D. L., FRANCKE, U. 1999; 56 (3): 357-358

    View details for Web of Science ID 000079390000018

    View details for PubMedID 10087207

  • TBL2, a novel transducin family member in the WBS deletion: characterization of the complete sequence, genomic structure, transcriptional variants and the mouse ortholog CYTOGENETICS AND CELL GENETICS Jurado, L. A., Wang, Y. K., FRANCKE, U., Cruces, J. 1999; 86 (3-4): 277-284

    Abstract

    Williams-Beuren syndrome (WBS) is a developmental disorder with multi-system manifestations caused by haploinsufficiency for contiguous genes deleted in chromosome region 7q11.23. The size of the deletion is similar in most patients due to a genomic duplication that predisposes to unequal meiotic crossover events. While hemizygosity at the elastin locus is responsible for the cardiovascular features, the contribution of other genes to the WBS phenotype remains to be demonstrated. We have identified a novel gene, TBL2, in the common WBS deletion. TBL2 is expressed as a 2. 4-kb transcript predominantly in testis, skeletal muscle, heart and some endocrine tissues, with a larger approximately 5-kb transcript detected ubiquitously at lower levels. TBL2 encodes a protein with four putative WD40-repeats. An alternatively spliced transcript in TBL2 introduces a novel second exon with an in frame stop codon. This mRNA encodes a 75 amino acid protein with 43 amino acids identical to TBL2 at the N-terminus and no known functional domain. The mouse homolog, Tbl2, shows 84% sequence identity at the nucleotide level and 92% similarity at the amino acid level. Comparison of the mouse and human sequences identifies a conserved region that extends upstream of the previously published sequence with an initiation codon common to both species that adds 21 amino acids at the N-terminus. The Tbl2 gene has been mapped to mouse chromosome 5 in a region of conserved synteny with human 7q11.23. Since haploinsufficiency has been shown for other WD-repeat containing proteins, hemizygosity of TBL2 may contribute to some of the aspects of the complex WBS phenotype.

    View details for Web of Science ID 000083951300023

    View details for PubMedID 10575226

  • Williams-Beuren syndrome: genes and mechanisms HUMAN MOLECULAR GENETICS FRANCKE, U. 1999; 8 (10): 1947-1954

    Abstract

    Williams-Beuren syndrome (WBS; OMIM 194050) is caused by heterozygous deletions of approximately 1.6 Mb of chromosomal sub-band 7q11.23. The deletions are rather uniform in size as they arise spontaneously by inter- or intrachromosomal crossover events within misaligned duplicated regions of high sequence identity that flank the typical deletion. This review will discuss the status of the molecular characterization of the deletion and flanking regions, the genes identified in the deletion region and their possible roles in generating the complex multi-system clinical phenotype.

    View details for Web of Science ID 000082952800016

    View details for PubMedID 10469848

  • Identification of GTF2IRD 1, a putative transcription factor within the Williams-Beuren syndrome deletion at 7q11.23 CYTOGENETICS AND CELL GENETICS Franke, Y., Peoples, R. J., FRANCKE, U. 1999; 86 (3-4): 296-304

    Abstract

    Williams-Beuren syndrome (WBS) is a microdeletion syndrome caused by haploinsufficiency of genes at 7q11.23. Here we describe the identification and characterization of a novel gene named GTF2IRD1, for GTF2I-repeat domain 1, within the WBS deletion region. Northern blot analysis revealed ubiquitous expression during development with two transcripts of 3.6 kb and 5.0 kb generated by alternative splicing. GTF2IRD1 encodes a protein of 944 amino acids that contains a region of high similarity to a unique motif with helix-loop-helix forming potential occurring within the transcription factor GTF2I. Analogous to TFII-I, the product of GTF2IRD1 may have the ability to interact with other HLH-proteins and function as a transcription factor or as a negative transcriptional regulator. A recent report of the identification of a muscle-specific transcription factor, MusTRD1, supports this hypothesis (O'Mahoney et al., 1998). The open reading frame described for MusTRD1 is identical to that of GTF2IRD1; however, the putative MusTRD1-protein is 486 amino acids shorter than the predicted protein encoded by GTF2IRD1. A heterozygous deletion of GTF2IRD1 may contribute to the complex WBS phenotype.

    View details for Web of Science ID 000083951300026

    View details for PubMedID 10575229

  • Novel mutations in the Wiskott-Aldrich syndrome protein gene and their effects on transcriptional, translational, and clinical phenotypes HUMAN MUTATION Lemahieu, V., Gastier, J. M., FRANCKE, U. 1999; 14 (1): 54-66

    Abstract

    Wiskott-Aldrich syndrome (WAS) is an X-linked recessive immunodeficiency characterized by thrombocytopenia, eczema, and recurrent infections, and caused by mutations in the WAS protein (WASP) gene. WASP contains several functional domains through which it interacts with proteins involved in intracellular signaling and regulation of the actin cytoskeleton. In this report, 17 WASP gene mutations were identified, 12 of which are novel. DNA of affected males and obligate carriers was PCR amplified and analyzed by SSCA, heteroduplex analysis, and direct sequencing. The effects of the mutations at the mRNA and protein level were ascertained by RT-PCR and Western blot analyses. All missense mutations were located in exons 1-4. Most of the nonsense, frameshift and splice site mutations were found in exons 6-11. Mutations that alter splice sites led to the synthesis of several types of mRNAs, a fraction of which represented the normally spliced product. The presence of normally spliced transcripts was correlated with a milder phenotype. When one such case was studied by Western blotting, reduced amounts of normal-size WASP were present. In other cases as well, a correlation was found between the amount of normal or mutant WASP present and the phenotypes of the affected individuals. No protein was detected in two individuals with severe WAS. Reduced levels of a normal-size WASP with a missense mutation were seen in two individuals with XLT. It is concluded that mutation analysis at the DNA level is not sufficient for predicting clinical course. Studies at the transcript and protein level are needed for a better assessment.

    View details for Web of Science ID 000081207100007

    View details for PubMedID 10447259

  • Conserved chromosomal location and genomic structure of human and mouse fatty-acid amide hydrolase genes and evaluation of clasper as a candidate neurological mutation GENOMICS Wan, M. H., Cravatt, B. F., Ring, H. J., Zhang, X. Y., FRANCKE, U. 1998; 54 (3): 408-414

    Abstract

    Fatty-acid amide hydrolase (FAAH) is a membrane-bound enzyme that degrades neuromodulatory fatty acid amides, such as oleamide and anandamide, and is expressed in the mammalian central nervous system. To evaluate FAAH genes as candidates for neurogenetic diseases in humans and mice, we have mapped the loci in both species and have determined their intron-exon structures. The human FAAH gene was mapped to region 1p34-p35, closely linked to D1S197 and D1S443, by using PCR analysis of somatic cell hybrid (SCH) and radiation hybrid mapping panels. Analysis of an SCH mapping panel and a mouse interspecific backcross panel has localized the Faah gene to the conserved syntenic region on mouse chromosome 4, close to the neurological mutation clasper. Faah gene rearrangements were excluded by Southern blot analysis of clasper DNA. No sequence abnormality was detected in PCR products containing the 15 exons and splice junctions of the mouse Faah gene. FAAH protein levels were normal in clasper mouse tissues as determined by enzyme activity assays and Western blotting.

    View details for Web of Science ID 000077932000005

    View details for PubMedID 9878243

  • Genes for the CPE receptor (CPETR1) and the human homolog of RVP1 (CPETR2) are localized within the Williams-Beuren syndrome deletion GENOMICS Paperna, T., Peoples, R., Wang, Y. K., Kaplan, P., FRANCKE, U. 1998; 54 (3): 453-459

    Abstract

    Williams-Beuren syndrome (WBS) is a neurodevelopmental disorder affecting multiple systems. Haploinsufficiency of genes deleted in chromosomal region 7q11.23 is the likely cause for this syndrome. We now report the localization of the genes for the CPE-R (Clostridium perfringens enterotoxin receptor, CPETR1) and the human homolog of RVP1 (rat ventral prostate 1 protein, CPETR2), both previously mapped to 7q11, to the WBS critical region. A single nucleotide polymorphism (SNP) present in CPETR1 has been identified and was used to determine parental origin of the deleted allele in five informative families. The mouse homologs Cpetr1 and Cpetr2 were identified and mapped to the conserved syntenic region on mouse chromosome 5. Northern blot analysis of CPETR1 demonstrates tissue specificity, with expression in kidney, lung, thyroid, and gastrointestinal tissues. In mouse, Cpetr1 is expressed in the early embryo, appears to be developmentally upregulated during gestation, and is present in adult tissues. Our results suggest a role for CPE-R in internal organ development and function during pre- and postnatal life.

    View details for Web of Science ID 000077932000010

    View details for PubMedID 9878248

  • Four contiguous amino acid substitutions, identified in patients with Laron syndrome, differently affect the binding affinity and intracellular trafficking of the growth hormone receptor JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM Wojcik, R., Berg, M. A., Esposito, N., Geffner, M. E., Sakati, N., Reiter, E. O., Dower, S., FRANCKE, U., Postel-Vinay, M. C., Finidori, J. 1998; 83 (12): 4481-4489

    Abstract

    We have analyzed the GH receptor (GHR) gene in four individuals with Laron syndrome, and a missense mutation was identified for each patient in the extracellular domain of the GHR (D152H, I153T, Q154P, and V155G). The D152H mutation was previously reported. We have reproduced the three novel mutations in the GHR complementary DNA and analyzed their consequences in human 293 transfected cells. In cells expressing the I153T and V155G mutants, binding of [125I]human GH at the cell surface was very low, whereas binding to total membrane fractions was much less affected, suggesting impaired cell surface expression. Binding assays with cells expressing the Q154P mutant revealed severe defects both at the cell surface and in total particulate membrane fractions. Immunofluorescence experiments confirmed that cell surface expression of the three mutants was altered, and colocalization studies suggested that most of the mutant receptors are retained in the endoplasmic reticulum. Endoglycosidase H resistance tests also indicated that the majority of I153T and V155G GHRs are trapped in the endoplasmic reticulum. Thus, mutations on contiguous amino acids of the GHR result in various defects. The I153T, Q154P, and V155G mutations mainly affect intracellular trafficking and binding affinity of the receptor, whereas the D152H mutation affects receptor expression, dimerization, and signaling.

    View details for Web of Science ID 000077359400052

    View details for PubMedID 9851797

  • Clinical objectives in medical genetics for undergraduate medical students GENETICS IN MEDICINE Friedman, J. M., Blitzer, M., Elsas, L. J., FRANCKE, U., WILLARD, H. F. 1998; 1 (1): 54-55

    View details for Web of Science ID 000082704300013

    View details for PubMedID 11261431

  • Five SWI/SNF-related, matrix-associated, actin-dependent regulator of chromatin (SMARC) genes are dispersed in the human genome GENOMICS Ring, H. Z., Vameghi-Meyers, V., Wang, W. D., Crabtree, G. R., FRANCKE, U. 1998; 51 (1): 140-143

    Abstract

    The SWI/SNF-related, matrix-associated, actin-dependent regulators of chromatin (SMARC), also called BRG1-associated factors, are components of human SWI/SNF-like chromatin-remodeling protein complexes. We mapped five human SMARC genes toregions on four different human chromosomes, SMARCC1 to 3p23-p21, SMARCC2 to 12q13-q14, SMARCD1 to 12q13-q14, SMARCD2 to 17q23-q24, and SMARCD3 to 7q35-q36. SMARCC1, SMARCC2, and SMARCD1 are assigned to chromosomal regions that are frequently involved in somatic rearrangements in human cancers. SMARCD1 was mapped to the critical region of Allgrove syndrome; however, no mutation was identified in one Allgrove syndrome family studied.

    View details for Web of Science ID 000075182000018

    View details for PubMedID 9693044

  • A single mutation that results in an Asp to His substitution and partial exon skipping in a family with congenital contractural arachnodactyly HUMAN GENETICS Babcock, D., Gasner, C., FRANCKE, U., Maslen, C. 1998; 103 (1): 22-28

    Abstract

    Congenital contractural arachnodactyly (CCA) is an autosomal dominant disorder of connective tissue and is characterized by multiple congenital contractures, arachnodactyly, and external ear malformations. Recent investigations indicate that mutations in the fibrillin-2 gene (FBN2) cause CCA. Here, we report a G-->C transversion at nucleotide 3340 (G3340C) of FBN2 in a family with phenotypic characteristics of CCA. The G3340C mutation predicts the substitution of histidine for aspartic acid at amino acid residue 1114 (Asp1114His) and also alters the 5' donor splice site consensus sequence of exon 25. Reverse transcription/polymerase chain reaction and DNA sequence analyses demonstrate that this missense mutation also causes low level in-frame mis-splicing of exon 25 (del exon 25). Consequently, this single point mutation produces a heterogeneous population of mutant fibrillin-2 molecules in a single individual. Despite the complex manifestation of the mutation, it is associated with a relatively mild phenotype. Analysis of FBN2 allele expression in cultured dermal fibroblasts derived from the proband has shown that the mutant allele is preferentially expressed, contributing about 84% of the total transcript. This indicates that an overabundance of mutant transcript does not necessarily correlate with a more severe CCA phenotype.

    View details for PubMedID 9737771

  • Stature in Ecuadorians heterozygous for growth hormone receptor gene E180 splice mutation does not differ from that of homozygous normal relatives JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM Rosenbloom, A. L., Guevara-Aguirre, J., Berg, M. A., FRANCKE, U. 1998; 83 (7): 2373-2375

    Abstract

    Heterozygosity for certain mutations of the GH receptor (GHR) gene has been proposed as the cause of partial resistance to GH, and there has been a recent demonstration of a dominant-negative effect of such a mutation in a mother and child. To examine the effect of heterozygosity in a large genetically homogeneous population with GHR deficiency, in which a substantial number of heterozygous (carrier) subjects and homozygous normal individuals can be compared, we studied a population in Ecuador in which 70 individuals with GHR deficiency were homozygous for the E180 splice mutation. We found that 58 heterozygous relatives of probands were not significantly shorter than 37 homozygous normal relatives [SD score (SDS) for height -1.85 +/- 1.04 (SD) vs. -1.55 +/- 0.96, P > 0.10]. When only those families with both homozygous normals and carriers were compared, the 33 heterozygous and 29 normal relatives did not differ significantly in height SDS (-1.98 +/- 1.07 vs. -1.77 +/- 0.91, P > 0.3). If heterozygosity for the E180 splice mutation were to influence stature, heights of heterozygous parents of probands would be expected to correlate with those of probands and of carriers who are their offspring and not with heights of their homozygous normal children. Parental height SDS did not correlate with height SDS of affected offspring (r = 0.24). For unaffected siblings as a group or analyzed separately as normals or carriers, there was a strong correlation between parental and offspring SDS for height (P < 0.01 for all comparisons). Thus, the effect of homozygosity for the GHR mutation was so profound as to abolish parental influence on height, and there was no difference in the influence of parental stature between carrier and noncarrier offspring. These findings demonstrate no meaningful effect on stature of heterozygosity for the E180 splice mutation of the GHR, which is a functional null mutation and, in the homozygous state, results in profound short stature from severe insulin-like growth factor-I deficiency.

    View details for Web of Science ID 000074562200025

    View details for PubMedID 9661611

  • A severely affected male born into a Rett syndrome kindred supports X-linked inheritance and allows extension of the exclusion map AMERICAN JOURNAL OF HUMAN GENETICS Schanen, C., FRANCKE, U. 1998; 63 (1): 267-269

    View details for Web of Science ID 000074761000035

    View details for PubMedID 9637791

  • p48 activates a UV-damaged-DNA binding factor and is defective in xeroderma pigmentosum group E cells that lack binding activity MOLECULAR AND CELLULAR BIOLOGY Hwang, B. J., Toering, S., FRANCKE, U., Chu, G. 1998; 18 (7): 4391-4399

    Abstract

    A subset of xeroderma pigmentosum (XP) group E cells lack a factor that binds to DNA damaged by UV radiation. This factor can be purified to homogeneity as p125, a 125-kDa polypeptide. However, when cDNA encoding p125 is translated in vitro, only a small fraction binds to UV-damaged DNA, suggesting that a second factor is required for the activation of p125. We discovered that most hamster cell lines expressed inactive p125, which was activated in somatic cell hybrids containing human chromosome region 11p11.2-11cen. This region excluded p125 but included p48, which encodes a 48-kDa polypeptide known to copurify with p125 under some conditions. Expression of human p48 activated p125 binding in hamster cells and increased p125 binding in human cells. No such effects were observed from expression of p48 containing single amino acid substitutions from XP group E cells that lacked binding activity, demonstrating that the p48 gene is defective in those cells. Activation of p125 occurred by a "hit-and-run" mechanism, since the presence of p48 was not required for subsequent binding. Nevertheless, p48 was capable of forming a complex with p125 either bound to UV-damaged DNA or in free solution. It is notable that hamster cells fail to efficiently repair cyclobutane pyrimidine dimers in nontranscribed DNA and fail to express p48, which contains a WD motif with homology to proteins that reorganize chromatin. We propose that p48 plays a role in repairing lesions that would otherwise remain inaccessible in nontranscribed chromatin.

    View details for Web of Science ID 000074380100077

    View details for PubMedID 9632823

  • Evaluation of two X chromosomal candidate genes for Rett syndrome: Glutamate dehydrogenase-2 (GLUD2) and rab GDP-dissociation inhibitor (GDI1) AMERICAN JOURNAL OF MEDICAL GENETICS Wan, M. H., FRANCKE, U. 1998; 78 (2): 169-172

    Abstract

    The most likely cause of the Rett syndrome (RTT) is an X-linked dominant mutation lethal in hemizygous males. Previous exclusion mapping studies have identified putative regions for the RTT gene on the X chromosome. In the present study, we evaluated two candidate genes, glutamate dehydrogenase-2 (GLUD2) and rab GDP-dissociation inhibitor (GDI1/XAP-4), chosen because of their expression patterns and functions in the central nervous system and their location in the nonexcluded region of Xq. The intronless gene GLUD2, located in Xq25 and expressed in neuronal and testicular tissues, is involved in the metabolism of glutamate, a neurotransmitter reported to be elevated in the spinal fluid of RTT individuals. The GLUD2 gene was screened for mutations by Southern hybridization and by direct sequencing of polymerase chain reaction (PCR) products. The GDI1 gene in Xq28, also known as RABGDIA or XAP-4, encodes a human GDI that is expressed predominantly in neuronal and sensory tissues. All 11 exons and splice junctions of the GDI1 gene were PCR-amplified and sequenced directly or screened by single-strand conformation analysis. No mutation in either of these two genes was found in 22 RTT patients. Therefore, GLUD2 and GDI1 can be excluded as candidate genes for this syndrome.

    View details for Web of Science ID 000074511200014

    View details for PubMedID 9674910

  • Exclusion of the gastrin-releasing peptide receptor (GRPR) locus as a candidate gene for Rett syndrome AMERICAN JOURNAL OF MEDICAL GENETICS Heidary, G., HAMPTON, L. L., Schanen, N. C., Rivkin, M. J., DARRAS, B. T., Battey, J., FRANCKE, U. 1998; 78 (2): 173-175

    Abstract

    The gene for the gastrin-releasing peptide receptor (GRPR) has been mapped to a candidate region for Rett syndrome (RTT) on the short arm of the X chromosome. The recent report of a translocation that disrupted the gene in an individual with mental retardation and autistic behavior prompted us to examine GRPR as a possible locus for RTT. Genomic polymerase chain reaction amplification of exons followed by single-strand conformation analysis screening in 25 unrelated RTT-affected individuals and by direct sequencing in 12 others has failed to detect any mutation. No gross structural rearrangements were found by Southern analysis of DNA from six unrelated RTT-affected individuals. A high-frequency biallelic polymorphism caused by two single nucleotide substitutions in exon 2 was discovered. The allele frequencies were identical in the RTT population as compared to 100 normal control X chromosomes. This polymorphism will enable future evaluation of the GRPR locus as a candidate for other X-linked mental retardation or neurobehavioral syndromes.

    View details for Web of Science ID 000074511200015

    View details for PubMedID 9674911

  • Retraction. The TSG101 tumor susceptibility gene is located in chromosome 11 band p15 and is mutated in human breast cancer. Cell Li, L., FRANCKE, U., Cohen, S. N. 1998; 93 (4): following 660-?

    View details for PubMedID 9867424

  • A mouse model for Prader-Willi syndrome imprinting-centre mutations NATURE GENETICS Yang, T., Adamson, T. E., Resnick, J. L., Leff, S., Wevrick, R., FRANCKE, U., Jenkins, N. A., Copeland, N. G., Brannan, C. I. 1998; 19 (1): 25-31

    Abstract

    Imprinting in the 15q11-q13 region involves an 'imprinting centre' (IC), mapping in part to the promoter and first exon of SNRPN. Deletion of this IC abolishes local paternally derived gene expression and results in Prader-Willi syndrome (PWS). We have created two deletion mutations in mice to understand PWS and the mechanism of this IC. Mice harbouring an intragenic deletion in Snrpn are phenotypically normal, suggesting that mutations of SNRPN are not sufficient to induce PWS. Mice with a larger deletion involving both Snrpn and the putative PWS-IC lack expression of the imprinted genes Zfp127 (mouse homologue of ZNF127), Ndn and Ipw, and manifest several phenotypes common to PWS infants. These data demonstrate that both the position of the IC and its role in the coordinate expression of genes is conserved between mouse and human, and indicate that the mouse is a suitable model system in which to investigate the molecular mechanisms of imprinting in this region of the genome.

    View details for Web of Science ID 000073346700012

    View details for PubMedID 9590284

  • Structure of the human paralemmin gene (PALM), mapping to human chromosome 19p13.3 and mouse chromosome 10, and exclusion of coding mutations in grizzled, mocha, jittery, and hesitant mice GENOMICS Burwinkel, B., Miglierini, G., Jenne, D. E., Gilbert, D. J., Copeland, N. G., Jenkins, N. A., Ring, H. Z., FRANCKE, U., Kilimann, M. W. 1998; 49 (3): 462-466

    Abstract

    Paralemmin is a newly identified protein that is associated with the plasma membrane and with intracellular membranes through a lipid anchor. It is abundant in brain, is expressed at intermediate levels in the kidney and in endocrine cells, and occurs at low levels in many other tissues. As it is a candidate for genetic disorders that affect membrane functions, we have determined the structure of the human paralemmin gene, PALM, showing that it is organized into nine exons. Moreover, we have performed chromosomal assignments of the human and mouse paralemmin genes, localizing them to regions of homology at human 19p13.3 and the central mouse chromosome 10. Finally, mutation analysis using RNA from mice homozygous for the mutant genes grizzled (gr), mocha (mh), mocha 2J (mh2J), jittery (ji) and hesitant (ji(hes)), which map to this area, excluded mutations in their Palm coding sequences.

    View details for Web of Science ID 000073834000017

    View details for PubMedID 9615234

  • Toso, a cell surface, specific regulator of Fas-induced apoptosis in T cells IMMUNITY Hitoshi, Y., Lorens, J., Kitada, S. I., Fisher, J., Labarge, M., Ring, H. Z., FRANCKE, U., Reed, J. C., Kinoshita, S., Nolan, G. P. 1998; 8 (4): 461-471

    Abstract

    Fas is a surface receptor that can transmit signals for apoptosis. Using retroviral cDNA library-based functional cloning we identified a gene, toso, that blocks Fas-mediated apoptosis. Toso expression was confined to lymphoid cells and was enhanced after cell-specific activation processes in T cells. Toso appeared limited to inhibition of apoptosis mediated by members of the TNF receptor family and was capable of inhibiting T cell self-killing induced by TCR activation processes that up-regulate Fas ligand. We mapped the effect of Toso to inhibition of caspase-8 processing, the most upstream caspase activity in Fas-mediated signaling, potentially through activation of cFLIP. Toso therefore serves as a novel regulator of Fas-mediated apoptosis and may act as a regulator of cell fate in T cells and other hematopoietic lineages.

    View details for Web of Science ID 000073348000008

    View details for PubMedID 9586636

  • A duplicated gene in the breakpoint regions of the 7q11.23 Williams-Beuren syndrome deletion encodes the initiator binding protein TFII-I and BAP-135, a phosphorylation target of BTK HUMAN MOLECULAR GENETICS Jurado, L. A., Wang, Y. K., Peoples, R., Coloma, A., Cruces, J., Francke, U. 1998; 7 (3): 325-334

    Abstract

    Williams-Beuren syndrome (WBS) is a neurodevelopmental disorder with multisystemic manifestations caused by heterozygosity for a partial deletion of chromosome band 7q11.23. The breakpoints cluster within regions located approximately 1 cM either side of the elastin (ELN) locus. We have characterized a duplicated region near the common deletion breakpoints, which includes a transcribed gene. The centromeric (C) and telomeric (T) copies are almost identical in the duplicated 3[prime] portions but diverge at their 5[prime]-ends. C-specific 4.3 kb mRNA and T-specific 5.4 kb mRNA are widely expressed in embryonic and adult tissues. The telomeric gene gives rise to several alternatively spliced forms and is deleted in all WBS individuals who have documented ELN deletions. Database searches revealed that this gene encodes BAP-135, a protein phosphorylated by Bruton's tyrosine kinase in B cells, as well as the multifunctional transcription factor TFII-I, hence the gene name GTF2I. The centromeric gene is not deleted in WBS and appears to be a partially truncated expressed pseudogene with no protein product (gene name GTF2IP1). Both loci map to different genomic clone contigs that also contain other deleted and non-deleted loci. A probe from the shared region recognizes a >3 Mb Not I junction fragment that is unique to individuals with the WBS deletion. Therefore, the duplicated region containing GTF2I and GTF2IP1 respectively is located close to the deletion breakpoints and may predispose to unequal meiotic recombination between chromosome 7 homologs and/or to intrachromosomal rearrangements. Hemizygosity for GTF2I may also contribute to the WBS phenotype.

    View details for Web of Science ID 000072513800001

    View details for PubMedID 9466987

  • A mouse single-copy gene, Gtf2i, the homolog of human GTF2I, that is duplicated in the Williams-Beuren syndrome deletion region GENOMICS Wang, Y. K., Perez-Jurado, L. A., FRANCKE, U. 1998; 48 (2): 163-170

    Abstract

    We have cloned and characterized Gtf2i, the mouse homolog of human GTF2I (general transcription factor II-I), which encodes BAP-135, a target for Bruton's tyrosine kinase. GTF2I represents the telomeric and functional copy of a duplicated gene flanking the 2-Mb Williams-Beuren syndrome (WBS) common deletion at 7q11.23. GTF2I is deleted in WBS, while a truncated centromeric pseudogene (GTF2IP1) is not deleted. In mouse, there appears to be only a single locus, Gtf2i, which we mapped to mouse chromosome 5 in a region of conserved mouse-human synteny. Gtf2i is 87.7% identical to GTF2I at the nucleotide and 97% at the amino acid level and generates several alternatively spliced transcripts. The gene is widely expressed in adult tissues and equally in all areas of the brain. Gtf2i transcript is detectable in ES cells by RT-PCR and on Northern blots of tissues from 7-dpc embryos. A ubiquitous expression pattern is seen by Northern and tissue in situ hybridization studies of 14-dpc embryos.

    View details for Web of Science ID 000072674100003

    View details for PubMedID 9521869

  • Marfan Database (third edition): new mutations and new routines for the software NUCLEIC ACIDS RESEARCH Collod-Beroud, G., Beroud, C., Ades, L., Black, C., Boxer, M., Brocks, D. J., Holman, K. J., De Paepe, A., FRANCKE, U., Grau, U., Hayward, C., Klein, H. G., Liu, W. G., Nuytinck, L., Peltonen, L., Perez, A. B., Rantamaki, T., JUNIEN, C., Boileau, C. 1998; 26 (1): 229-233

    Abstract

    The Marfan database is a software that contains routines for the analysis of mutations identified in the FBN1 gene that encodes fibrillin-1. Mutations in this gene are associated not only with Marfan syndrome but also with a spectrum of overlapping disorders. The third version of the Marfan database contains 137 entries. The software has been modified to accommodate four new routines and is now accessible on the World Wide Web at http://www.umd.necker.fr

    View details for Web of Science ID 000071778900054

    View details for PubMedID 9399842

  • Imprinted genes in the Prader-Willi deletion - Discussion Symposium on Epigenetics Jaenisch, Francke, Barlow, Riggs, Laird, Gasser, Surani, Bird, Wolffe, Martienssen, Paro, Bestor, Reik, Matzke JOHN WILEY & SONS LTD. 1998: 275–279
  • Gene silencing by methyl-CpG-binding proteins - Discussion Symposium on Epigenetics Barlow, Bird, Jaenisch, Wolffe, Gasser, Horz, Finnegan, Allshire, Fedoroff, Lyon, Francke, Flavell JOHN WILEY & SONS LTD. 1998: 16–21
  • The dynamics of globin gene expression and position effects - Discussion Symposium on Epigenetics Bird, Grosveld, Barlow, Wolffe, Jaenisch, Gasser, Riggs, Horz, Francke, Finnegan, Paro, Laird, Pillus, Lyon JOHN WILEY & SONS LTD. 1998: 79–86
  • Heritable chromatin states induced by the Polycomb and trithorax group genes - Discussion Symposium on Epigenetics Fedoroff, Paro, Wolffe, Horz, Riggs, Laird, Reik, Allshire, Finnegan, Barlow, Jaenisch, Francke, Wilkins, Grosveld, Gasser JOHN WILEY & SONS LTD. 1998: 61–66
  • Identification of the WBSCR9 gene, encoding a novel transcriptional regulator, in the Williams-Beuren syndrome deletion at 7q11.23 CYTOGENETICS AND CELL GENETICS Peoples, R. J., Cisco, M. J., Kaplan, P., Francke, U. 1998; 82 (3-4): 238-246

    Abstract

    We have identified a novel gene (WBSCR9) within the common Williams-Beuren syndrome (WBS) deletion by interspecies sequence conservation. The WBSCR9 gene encodes a roughly 7-kb transcript with an open reading frame of 1483 amino acids and a predicted protein product size of 170.8 kDa. WBSCR9 is comprised of at least 20 exons extending over 60 kb. The transcript is expressed ubiquitously throughout development and is subject to alternative splicing. Functional motifs identified by sequence homology searches include a bromodomain; a PHD, or C4HC3, finger; several putative nuclear localization signals; four nuclear receptor binding motifs; a polyglutamate stretch and two PEST sequences. Bromodomains, PHD motifs and nuclear receptor binding motifs are cardinal features of proteins that are involved in chromatin remodeling and modulation of transcription. Haploinsufficiency for WBSCR9 gene products may contribute to the complex phenotype of WBS by interacting with tissue-specific regulatory factors during development.

    View details for Web of Science ID 000077796000023

    View details for PubMedID 9858827

  • Imprinted genes in the Prader-Willi deletion Symposium on Epigenetics FRANCKE, U. JOHN WILEY & SONS LTD. 1998: 264–275

    Abstract

    Parent-of-origin-specific deletions of proximal chromosome 15q cause either the Prader-Willi syndrome (paternal deletion) or the Angelman syndrome (maternal deletion), two distinct neurodevelopmental disorders. In contrast to the Angelman syndrome, which can also be caused by mutations in a single gene (UBE3A, encoding a ubiquitin ligase), the Prader-Willi syndrome is caused by deletions in about two-thirds of cases and by maternal uniparental disomy in the remaining third. The consequence of both mechanisms, in addition to rare microdeletions or so-called 'imprinting mutations', is lack of the products of multiple genes in the region that are normally expressed only from the paternal chromosome. One gene that is consistently silent in the Prader-Willi syndrome is SNRPN, which encodes the small nuclear ribonucleoprotein particle-associated polypeptide N that forms part of the spliceosomes in the brain. A systematic search for other imprinted genes in the Prader-Willi syndrome region revealed a paternally expressed transcript (IPW, for imprinted in the Prader-Willi region) and a similarly imprinted mouse homologue (Ipw) in the conserved syntenic region on mouse chromosome 7. Ipw is highly expressed in the brain and alternatively spliced to generate different transcripts. Since there is no open reading frame that is conserved in the human and mouse IPW genes, they are postulated to function as untranslated RNAs, possibly regulating transcription in cis in the region.

    View details for Web of Science ID 000076284200034

    View details for PubMedID 9601023

  • Localization of the human HIP1 gene close to the elastin (ELN) locus on 7q11.23 GENOMICS Wedemeyer, N., Peoples, R., Himmelbauer, H., Lehrach, H., FRANCKE, U., Wanker, E. E. 1997; 46 (2): 313-315

    View details for Web of Science ID A1997YL01600022

    View details for PubMedID 9417924

  • Mouse latent TGF-beta binding protein 2: molecular cloning and developmental expression BIOCHIMICA ET BIOPHYSICA ACTA-GENE STRUCTURE AND EXPRESSION Fang, J. M., Li, X., Smiley, E., FRANCKE, U., Mecham, R. P., Bonadio, J. 1997; 1354 (3): 219-230

    Abstract

    The molecular cloning and developmental expression of mouse LTBP-2 are presented here. We established the identity of the cDNA by sequence comparison (80% identity with human LTBP-2) and by chromosome localization (mouse chromosome 12, band D, a region of conserved synteny with the human LTBP-2 gene). In contrast to LTBP-1 and LTBP-3, mouse LTBP-2 apparently is a more modular protein, with proline/glycine-rich sequences always alternating with clusters of cysteine-rich structural motifs. We found for the first time that LTBP-2 gene expression in mouse embryos was restricted to cartilage perichondrium and blood vessels, a somewhat surprising result since other LTBP genes are widely expressed in rodent tissues. Therefore, mouse LTBP-2 may play a critical role in the assembly of latent TGF-beta complexes in developing elastic tissues such as cartilage and blood vessel.

    View details for Web of Science ID 000070986500005

    View details for PubMedID 9427531

  • Expression of human Wiskott-Aldrich syndrome protein in patients' primary hematopoietic cells and EBV-transduced cell lines after retroviral vector-mediated gene transduction. Huang, M. M., Wong, A., Weinberg, K. I., FRANCKE, U., Kohn, D. B. AMER SOC HEMATOLOGY. 1997: 1154–54
  • Identification of a gene (GPR30) with homology to the G-protein-coupled receptor superfamily associated with estrogen receptor expression in breast cancer GENOMICS Carmeci, C., Thompson, D. A., Ring, H. J., FRANCKE, U., Weigel, R. J. 1997; 45 (3): 607-617

    Abstract

    Using the technique of differential cDNA library screening, a cDNA clone was isolated from an estrogen receptor (ER)-positive breast carcinoma cell line (MCF7) cDNA library based upon the overexpression of this gene compared to an ER-negative cell line (MDA-MB-231). Sequence analysis of this clone determined that it shared significant homology to G-protein-coupled receptors. This receptor, GPCR-Br, was abundantly expressed in the ER-positive breast carcinoma cell lines MCF7, T-47D, and MDA-MB-361. Expression was absent or minimal in the ER-negative breast carcinoma cell lines BT-20, MDA-MB-231, and HBL-100. GPCR-Br was ubiquitously expressed in human tissues examined but was most abundant in placenta. GPCR-Br expression was examined in 11 primary breast carcinomas. GPCR-Br was detected in all 4 ER-positive tumors and only 1 of 7 ER-negative tumors. Based upon PCR analysis in hybrid cell lines, the gene for GPCR-Br (HGMW-approved symbol GPR30) was mapped to chromosome 7p22. The pattern of expression of GPCR-Br indicates that this receptor may be involved in physiologic responses specific to hormonally responsive tissues.

    View details for Web of Science ID A1997YF97300015

    View details for PubMedID 9367686

  • A new Rett syndrome family consistent with X-linked inheritance expands the X chromosome exclusion map AMERICAN JOURNAL OF HUMAN GENETICS Schanen, N. C., Dahle, E. J., Capozzoli, F., HOLM, V. A., Zoghbi, H. Y., FRANCKE, U. 1997; 61 (3): 634-641

    Abstract

    Although familial recurrences of Rett syndrome (RTT) comprise only approximately 1% of the reported cases, it is these cases that hold the key for the understanding of the genetic basis of the disorder. Families in which RTT occurs in mother and daughter, aunt and niece, and half sisters are consistent with dominant inheritance and variable expressivity of the phenotype. Recurrence of RTT in sisters is likely due to germ-line mosaicism in one of the parents, rather than to recessive inheritance. The exclusive occurrence of classic RTT in females led to the hypothesis that it is X-linked and may be lethal in males. In an X-linked dominant disorder, unaffected obligate-carrier females would be expected to show nonrandom or skewed inactivation of the X chromosome bearing the mutant allele. We investigated the X chromosome inactivation (XCI) patterns in the female members of a newly identified family with recurrence of RTT in a maternal aunt and a niece. Skewing of XCI is present in the obligate carrier in this family, supporting the hypothesis that RTT is an X-linked disorder. However, evaluation of the XCI pattern in the mother of affected half sisters shows random XCI, suggesting germ-line mosaicism as the cause of repeated transmission in this family. To determine which regions of the X chromosome were inherited concordantly/discordantly by the probands, we genotyped the individuals in the aunt-niece family and two previously reported pairs of half sisters. These combined exclusion-mapping data allow us to exclude the RTT locus from the interval between DXS1053 in Xp22.2 and DXS1222 in Xq22.3. This represents an extension of the previous exclusion map.

    View details for Web of Science ID A1997XY52300021

    View details for PubMedID 9326329

  • Silent mutation induces exon skipping of fibrillin-1 gene in Marfan syndrome NATURE GENETICS Liu, W. G., Qian, C. P., FRANCKE, U. 1997; 16 (4): 328-329

    View details for Web of Science ID A1997XM90900008

    View details for PubMedID 9241263

  • Ascending aortic aneurysm with or without features of Marfan syndrome and other fibrillinopathies: new insights. Seminars in thoracic and cardiovascular surgery FURTHMAYR, H., FRANCKE, U. 1997; 9 (3): 191-205

    Abstract

    More than 70 unique fibrillin-1 mutations have been identified in individuals with a variety of phenotypic changes. These range from severe neonatal lethal forms of Marfan syndrome to adult onset manifestations, mitral valve prolapse syndromes to isolated features such as ectopia lentis, Marfanoid body habitus and ascending aortic aneurysm and/or dissection. Fibrillin-1 mutations result in structurally and functionally defective fibrillin-1 molecules and microfibrils. Recent molecular genetic and fibrillin-1 biosynthesis studies suggest that individuals with fibrillin-1 abnormalities can be further subdivided into groups that are associated with distinct differences in severity and prognosis. In recognition of the expanding scope of related connective tissue disorders, we propose the terms microfibrillar disorder for disorders affecting fibrillin-containing microfibrils, and the more narrow concept of fibrillinopathy for clinical entities associated with abnormalities of fibrillin-1 or fibrillin-2. This latter category includes the previously defined disorders Marfan syndrome, congenital contractual arachnodactyly, and forms of ascending aortic aneurysm and/or dissection.

    View details for PubMedID 9263339

  • Is Rett syndrome caused by a triplet repeat expansion? NEUROPEDIATRICS Hofferbert, S., Schanen, N. C., Budden, S. S., FRANCKE, U. 1997; 28 (3): 179-183

    Abstract

    Rett syndrome is usually sporadic, but rare pedigrees with nonpenetrance in obligate carriers and possible anticipation suggest that it could be caused by a triplet repeat expansion (TRE). Rett probands and controls were systematically screened for expansions of any of the 10 possible triplet repeats by using a modified Repeat Expansion Detection (RED) assay that had been shown to detect expanded disease alleles in myotonic dystrophy and Huntington disease. No significant expansions were found in 26 sporadic and six familial Rett probands. Our results exclude the possibility that Rett syndrome is caused by a large TRE. We cannot exclude, however, causation by a small TRE that is masked by the background of longer polymorphic repeats in the normal population.

    View details for Web of Science ID A1997XN48900008

    View details for PubMedID 9266557

  • The gene for microfibril-associated protein-1 (MFAP1) is located several megabases centromeric to FBN1 and is not mutated in Marfan syndrome HUMAN GENETICS Liu, W. G., Faraco, J., Qian, C. P., FRANCKE, U. 1997; 99 (5): 578-584

    Abstract

    Linkage studies have mapped the Marfan syndrome (MFS) locus to chromosome region 15q15-q21 with no convincing evidence of genetic heterogeneity. The fibrillin-1 (FBN1) gene, located at 15q21.1, that encodes the major component of the defective microfibrils, has been identified as the gene for MFS. However, extensive mutation screening in many laboratories has detected FBN1 mutations in only a fraction of MFS probands studied, leading to the hypothesis that the missing mutations could involve another microfibril gene located in the same region. Recently, the gene for microfibril-associated protein-1 (MFAP1, also called AMP) has been isolated and mapped to the 15q15-q21 region that overlaps the location of the FBN1 gene. Here we report that the two loci are physically close, making MFAP1 an alternative positional candidate gene for MFS. We have carried out MFAP1 mutation screening and gene expression analysis in 48 probands with MFS or related phenotypes who were selected for this study because their fibroblast cultures synthesized fibrillin at normal levels. No MFAP1 mutations were identified, and transcription occurred equally from both alleles. We conclude that the MFAP1 locus is not a reservoir for the hidden MFS mutations.

    View details for Web of Science ID A1997WW19000004

    View details for PubMedID 9150721

  • The pathogenicity of the Pro1148Ala substitution in the FBN1 gene: Causing or predisposing to Marfan syndrome and aortic aneurysm, or clinically innocent? HUMAN GENETICS Schrijver, I., Liu, W. G., FRANCKE, U. 1997; 99 (5): 607-611

    Abstract

    In individuals with the Marfan syndrome (MFS), mutations have been identified in the fibrillin-1 gene (FBN1) at 15q21.1. A proline-to-alanine change at position 1148 in exon 27 (Pro1148Ala) has been reported in probands with MFS, aortic aneurysm or Marfanoid-craniosynostosis. It was suggested that this mutation could be a risk factor for aortic dilatation, since it was rarely observed in control populations. To investigate further the pathogenicity of this substitution, we screened 416 unrelated control individuals by allele-specific oligonucleotide (ASO) hybridization. We found 16 individuals who carried the alanine allele (3.8%), 3 of whom were homozygous. Five were of Latin American and eight were of Asian extraction. We also screened 133 probands with MFS, aortic aneurysm or related connective tissue disorders and found 4 (3%) that were heterozygous for the 1148Ala allele. All positive results were confirmed by DNA sequencing. In 20 individuals with 1148Ala, we confirmed the association with the rarer A allele at the IVS27-5G-->A polymorphism. Our results suggest that the Pro1148Ala change is a polymorphism of ancient evolutionary origin that is more prevalent in Asian and Latin American than in Caucasian or African populations.

    View details for Web of Science ID A1997WW19000009

    View details for PubMedID 9150726

  • Molecular cloning, mapping to human chromosome 1 q21-q23, and cell binding characteristics of Sp alpha, a new member of the scavenger receptor cysteine-rich (SRCR) family of proteins JOURNAL OF BIOLOGICAL CHEMISTRY Gebe, J. A., Kiener, P. A., Ring, H. J., Li, X., FRANCKE, U., Aruffo, A. 1997; 272 (10): 6151-6158

    Abstract

    CD5 and CD6, two type I cell surface antigens predominantly expressed by T cells and a subset of B cells, have been shown to function as accessory molecules capable of modulating T cell activation. Here we report the cloning of a cDNA encoding Spalpha, a secreted protein that is highly homologous to CD5 and CD6. Spalpha has the same domain organization as the extracellular region of CD5 and CD6 and is composed of three SRCR (scavenger receptor cysteine rich) domains. Chromosomal mapping by fluorescence in situ hybridization and radiation hybrid panel analysis indicated that the gene encoding Spalpha is located on the long arm of human chromosome 1 at q21-q23 within contig WC1.17. RNA transcripts encoding Spalpha were found in human bone marrow, spleen, lymph node, thymus, and fetal liver but not in non-lymphoid tissues. Cell binding studies with an Spalpha immunoglobulin (Spalpha-mIg) fusion protein indicated that Spalpha is capable of binding to peripheral monocytes but not to T or B cells. Spalpha-mIg was also found to bind to the monocyte precursor cell lines K-562 and weakly to THP-1 but not to U937. Spalpha-mIg also bound to the B cell line Raji and weakly to the T cell line HUT-78. These findings indicate that Spalpha, a novel secreted protein produced in lymphoid tissues, may regulate monocyte activation, function, and/or survival.

    View details for Web of Science ID A1997WM64700011

    View details for PubMedID 9045627

  • A novel human homologue of the Drosophila frizzled wnt receptor gene binds wingless protein and is in the Williams syndrome deletion at 7q11.23 HUMAN MOLECULAR GENETICS Wang, Y. K., Samos, C. H., Peoples, R., PEREZJURADO, L. A., Nusse, R., FRANCKE, U. 1997; 6 (3): 465-472

    Abstract

    Williams syndrome (WS) is a developmental disorder with a characteristic personality and cognitive profile that is associated, in most cases, with a 2 Mb deletion of part of chromosome band 7q11.23. By applying CpG island cloning methods to cosmids from the deletion region, we have identified a new gene, called FZD3. Dosage blotting of DNA from 11 WS probands confirmed that it is located within the commonly deleted region. Sequence comparisons revealed that FZD3, encoding a 591 amino acid protein, is a novel member of a seven transmembrane domain receptor family that are mammalian homologs of the Drosophila tissue polarity gene frizzled. FZD3 is expressed predominantly in brain, testis, eye, skeletal muscle and kidney. Recently, frizzled has been identified as the receptor for the wingless (wg) protein in Drosophila. We show that Drosophila as well as human cells, when transfected with FZD3 expression constructs, bind Wg protein. In mouse, the wg homologous Wnt1 gene is involved in early development of a large domain of the central nervous system encompassing much of the midbrain and rostral metencephalon. The potential function of FZD3 in transmitting a Wnt protein signal in the human brain and other tissues suggests that heterozygous deletion of the FZD3 gene could contribute to the WS phenotype.

    View details for Web of Science ID A1997WN31600016

    View details for PubMedID 9147651

  • Molecular diagnosis and endocrine evaluation of a patient with a homozygous 7.0 kb deletion of the growth hormone (GH) gene cluster: Response to biosynthetic GH therapy JOURNAL OF PEDIATRIC ENDOCRINOLOGY & METABOLISM Jurado, L. A., Argente, J., Barrios, V., Pozo, J., Munoz, M. T., HERNANDEZ, M., FRANCKE, U. 1997; 10 (2): 185-190

    Abstract

    A significant proportion of cases of GH deficiency (5-30%) may be due to genetic causes. At least four Mendelian types of isolated GH deficiency (IGHD) have been delineated based on the mode of inheritance and the degree of GH deficiency, with IGHD type IA being the most severe. A 2 year-old girl, the second child of consanguineous parents, with short stature was diagnosed with IGHD type IA. The analysis of the genomic DNA of this patient, performed by polymerase chain reaction (PCR) amplification of the flanking regions of the GH-1 gene, showed a homozygous deletion of 7.0 kb of sequence including the GH-1 gene. She was treated with biosynthetic GH resulting in long-lasting catch-up growth during at least three years, despite a clinically irrelevant appearance of low binding capacity GH antibodies. Growth hormone-binding protein (GHBP) levels were normal at the time of diagnosis. In addition, GHBP plasma levels did not show any significant change during the three years of therapy with GH. Diagnosis of carrier status in family relatives was done by genotyping GH gene alleles by PCR amplification from blood spots on filter paper.

    View details for Web of Science ID A1997XA69900003

    View details for PubMedID 9364351

  • Backfoot, a novel homeobox gene, maps to human chromosome 5 (BFT) and mouse chromosome 13 (Bft) GENOMICS Shang, J., Li, X., Ring, H. Z., Clayton, D. A., FRANCKE, U. 1997; 40 (1): 108-113

    Abstract

    Homeobox genes play important roles in limb development. Backfoot is a recently identified mammalian homeobox gene whose temporal and spatial expression pattern during limb development suggests that it is a key component for specifying the identify and structure of the hindlimb. Here we report the chromosomal mapping of the Backfoot locus in human (BFT) and mouse (Bft). Using single-strand conformation analysis of PCR products amplified from a panel of somatic cell hybrid lines and two radiation hybrid (RH) panels, we have physically mapped BFT to human chromosome 5, closely linked to STS markers D5S2543, D5S458, D5S1947, and D5S1995 on the Stanford G3 RH map and to AFMA057VG5 and AFM350YB1 on the Gene-Bridge 4 RH map. Linkage analysis of a mouse inter-specific backcross panel (C57BL/6J x Mus musculus spretus) has localized Bft to the central part of mouse chromosome 13. The map position of Bft is near two mouse limb mutant loci defined as dumpy and mdac.

    View details for Web of Science ID A1997WJ33600014

    View details for PubMedID 9070926

  • An imprinted mouse transcript homologous to the human imprinted in Prader-Willi syndrome (IPW) gene HUMAN MOLECULAR GENETICS Wevrick, R., FRANCKE, U. 1997; 6 (2): 325-332

    Abstract

    The Prader-Willi syndrome (PWS) is caused by genomic alterations that inactivate imprinted, paternally expressed genes in human chromosome region 15q11-q13. IPW, a paternally expressed gene cloned from this region, is not expressed in individuals with PWS, and is thus a candidate for involvement in this disorder. The IPW transcript does not appear to encode a polypeptide, suggesting that it functions at the level of an RNA. We have now cloned a mouse gene, named Ipw, that has sequence similarity to a part of IPW and is located in the conserved homologous region of mouse chromosome 7. The Ipw cDNA also contains no long open reading frame, is alternatively spliced and contains multiple copies of a 147 bp repeat, arranged in a head-to-tail orientation, that are interrupted by the insertion of an intracisternal A particle sequence. Ipw is expressed predominantly in brain. In an interspecies (M.musculus x M.m.castaneus) F1 hybrid animal, expression of Ipw is limited to the paternal allele. We propose that Ipw is the murine homolog of IPW.

    View details for Web of Science ID A1997WH78900021

    View details for PubMedID 9063754

  • Molecular characterization of two mammalian bHLH-PAS domain proteins selectively expressed in the central nervous system PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Zhou, Y. D., Barnard, M., Tian, H., Li, X., Ring, H. Z., FRANCKE, U., Shelton, J., Richardson, J., Russell, D. W., McKnight, S. L. 1997; 94 (2): 713-718

    Abstract

    Here we describe two mammalian transcription factors selectively expressed in the central nervous system. Both proteins, neuronal PAS domain protein (NPAS) 1 and NPAS2, are members of the basic helix-loop-helix-PAS family of transcription factors. cDNAs encoding mouse and human forms of NPAS1 and NPAS2 have been isolated and sequenced. RNA blotting assays demonstrated the selective presence of NPAS1 and NPAS2 mRNAs in brain and spinal cord tissues of adult mice. NPAS1 mRNA was first detected at embryonic day 15 of mouse development, shortly after early organogenesis of the brain. NPAS2 mRNA was first detected during early postnatal development of the mouse brain. In situ hybridization assays using brain tissue of postnatal mice revealed an exclusively neuronal pattern of expression for NPAS1 and NPAS2 mRNAs. The human NPAS1 gene was mapped to chromosome 19q13.2-q13.3, and the mouse Npas1 gene to chromosome 7 at 2 centimorgans. Similarly, the human NPAS2 gene was assigned to chromosome 2p11.2-2q13, and the mouse Npas2 gene to chromosome 1 at 21-22 centimorgans. The chromosomal regions to which human NPAS1 and NPAS2 map are syntenic with those containing the mouse Npas1 and Npas2 genes, indicating that the mouse and human genes are true homologs.

    View details for Web of Science ID A1997WD88500062

    View details for PubMedID 9012850

  • The TSG101 tumor susceptibility gene is located in chromosome 11 band p15 and is mutated in human breast cancer CELL Li, L. M., Li, X., FRANCKE, U., Cohen, S. N. 1997; 88 (1): 143-154

    Abstract

    Recent work has identified a mouse gene (tsg101) whose inactivation in fibroblasts results in cellular transformation and the ability to produce metastatic tumors in nude mice. Here, we report that the human homolog, TSG101, which we isolated and mapped to chromosome 11, bands 15.1-15.2, a region proposed to contain tumor suppressor gene(s), is mutated at high frequency in human breast cancer. In 7 of 15 uncultured primary human breast carcinomas, intragenic deletions were shown in TSG101 genomic DNA and transcripts by gel and sequence analysis, and mutations affecting two TSG101 alleles were identified in four of these cancers. No TSG101 defects were found in matched normal breast tissue from the breast cancer patients. These findings strongly implicate TSG101 mutations in human breast cancer.

    View details for Web of Science ID A1997WC56900017

    View details for PubMedID 9019400

  • Denaturing HPLC-identified novel FBN1 mutations, polymorphisms, and sequence variants in Marfan syndrome and related connective tissue disorders GENETIC TESTING Liu, W. O., Oefner, P. J., Qian, C. P., Odom, R. S., FRANCKE, U. 1997; 1 (4): 237-242

    Abstract

    Marfan syndrome (MFS), a common connective tissue disorder, is caused by fibrillin-1 (FBN1) mutations that are scattered throughout the gene and are largely unique to individual families. Mutation detection in this large gene of 65 exons is a considerable technical challenge. To develop an efficient method capable of identifying all possible mutations in this gene, we have explored the use of a novel denaturing high-performance liquid chromatography (DHPLC) system. This technique compares two or more chromosomes as a mixture of denatured and reannealed PCR amplicons. Under partially denaturing conditions, heteroduplexes can be separated from homoduplexes. A panel of 94 DNA samples from individuals with MFS or related connective tissue disorders was screened exon-by-exon by this method. A total of 66 unique heteroduplex profiles was identified. Sequencing of the amplicons detected 37 novel and two previously reported mutations, as well as 15 novel and 10 known polymorphisms or unique sequence variants that are probably of no clinical significance. Of the 34 mutations found in definitive MFS cases, 16 were identified in the 21 samples that had not been screened before (76% detection rate) and 17/40 (43%) were in samples previously screened by other mutation detection methods. In 32 individuals with MFS-related phenotypes, five FBN1 mutations were identified (16%). Our results demonstrate the power of the DHPLC method to detect FBN1 mutations. It should be applicable for mutation screening in any gene in a large population.

    View details for Web of Science ID 000078308900002

    View details for PubMedID 10464652

  • Trinucleotide repeats in the human genome: Size distributions for all possible triplets and detection of expanded disease alleles in a group of Huntington disease individuals by the Repeat Expansion Detection method HUMAN MOLECULAR GENETICS Hofferbert, S., Schanen, N. C., Chehab, F., FRANCKE, U. 1997; 6 (1): 77-83

    Abstract

    Using a modified Repeat Expansion Detection (RED) assay, that was optimized for individual oligonucleotides, unrelated individuals were systematically screened for maximal repeat sizes of each of the ten possible trinucleotide repeats. Cloned trinucleotide repeats were generated and used as standards for the detectability of single copy trinucleotide repeat fragments. When the size distributions of trinucleotide repeats were compared to previously reported data, significant differences were found for the CTT repeat, which corresponds to the expanded GAA repeat in Friedreich ataxia, as well as for ATT, CCT and GTT repeats. Since 30-35% of normal individuals have CTG/CAG trinucleotide repeat sizes of 180 bp or more, we investigated the question whether small-scale CTG/CAG repeat expansions are detectable on a population basis by using the RED technique. We blindly screened 20 HD probands with CAG expansions of the HD gene, ranging in size between 120 and 174 bp, and found that a shift to larger CAG size ranges is clearly detectable when comparing the distribution of maximal repeat sizes in the disease group to a control group. Our study, therefore, demonstrates that the application of the RED assay to a population of probands and a population of controls allows the detection of small-scale CTG/CAG repeat expansions in the size range of the expanded HD gene and present in a single allele. We also provide standards and control data for the detection of other trinucleotide repeat expansions.

    View details for Web of Science ID A1997WD34900011

    View details for PubMedID 9002673

  • Structural, functional analysis and localization of the human CAP18 gene FEBS LETTERS Larrick, J. W., Lee, J., Ma, S. W., Li, X., FRANCKE, U., Wright, S. C., Balint, R. F. 1996; 398 (1): 74-80

    Abstract

    CAP18 is an antimicrobial protein found in specific granules of PMNs. The human CAP18 (HCAP18) gene was cloned from a human genomic phage library. Sequence analysis revealed the HCAP18 gene to have 4 exons spanning 3 kb, including 700 bp of upstream DNA. Using 3' RACE no homologs of human HCAP18 were found in human bone marrow or leukocyte populations. By PCR analysis of a somatic cell mapping panel and fluorescence in situ hybridization of a genomic clone to metaphase chromosomes the gene was mapped to chromosome band 3p21.3. Like several other genes expressed late in PMN development the CAP18 gene did not contain typical TATA box or CCAAT sequences. Expression in Cos 7 cells permitted limited mapping of the promoter function in upstream fragments of the HCAP18 gene. Western blot, Northern blot and RT-PCR analysis show HCAP18 to be produced specifically in granulocytes. This work forms the groundwork for future analysis of the genetic regulation of this antimicrobial protein during PMN differentiation.

    View details for Web of Science ID A1996VV87800014

    View details for PubMedID 8946956

  • Diagnostic test for the Prader-Willi syndrome by SNRPN expression in blood LANCET Wevrick, R., FRANCKE, U. 1996; 348 (9034): 1068-1069

    Abstract

    Prader-Willi syndrome (PWS) is caused by alterations of the paternally derived chromosome 15 or by maternal uniparental disomy. The gene for the small nuclear ribonucleoprotein polypeptide N (SNRPN) is expressed only from the paternally derived chromosome 15, due to epigenetic imprinting. The SNRPN gene is not expressed in any patients with PWS regardless of the underlying cytogenetic or molecular causes.To develop a rapid molecular diagnostic assay for PWS, we tested the expression of the SNRPN gene and a control gene in 9 patients with PWS and 40 control individuals by PCR analysis of reverse transcribed mRNA from blood leucocytes. We then tested 11 blood samples from patients with suspected PWS.SNRPN expression could readily be detected in blood leucocytes by PCR analysis in all control samples but not in samples from known PWS patients. Four suspected plus were negative for SNRPN expression were found to have chromosome 15 rearrangements, while the diagnosis of PWS was excluded in the remaining seven with normal SNRPN expression based on clinical, molecular, and cytogenetic findings.The SNRPN-expression test is rapid and reliable in the molecular diagnosis of Prader-Willi syndrome.

    View details for Web of Science ID A1996VM88600012

    View details for PubMedID 8874459

  • Molecular definition of the chromosome 7 deletion in Williams syndrome and parent-of-origin effects on growth AMERICAN JOURNAL OF HUMAN GENETICS Jurado, L. A., Peoples, R., Kaplan, P., Hamel, B. C., FRANCKE, U. 1996; 59 (4): 781-792

    Abstract

    Williams syndrome (WS) is a developmental disorder with variable phenotypic expression associated, in most cases, with a hemizygous deletion of part of chromosomal band 7q11.23 that includes the elastin gene (ELN). We have investigated the frequency and size of the deletions, determined the parental origin, and correlated the molecular results with the clinical findings in 65 WS patients. Hemizygosity at the ELN locus was established by typing of two intragenic polymorphisms, quantitative Southern analysis, and/or FISH. Polymorphic markers covering the deletion and flanking regions were ordered by a combination of genetic and physical mapping. Genotyping of WS patients and available parents for 13 polymorphisms revealed that of 65 clinically defined WS patients, 61 (94%) had a deletion of the ELN locus and were also hemizygous (or noninformative) at loci D7S489B, D7S2476, D7S613, D7S2472, and D7S1870. None of the four patients without ELN deletion was hemizygous at any of the polymorphic loci studied. All patients were heterozygous (or noninformative) for centromeric (D7S1816, D7S1483, and D7S653) and telomeric (D7S489A, D7S675, and D7S669) flanking loci. The genetic distance between the most-centromeric deleted locus, D7S489B, and the most-telomeric one, D7S1870, is 2 cM. The breakpoints cluster at approximately 1 cM to either side of ELN. In 39 families informative for parental origin, all deletions were de novo, and 18 were paternally and 21 maternally derived. Comparison of clinical data, collected in a standardized quantifiable format, revealed significantly more severe growth retardation and microcephaly in the maternal deletion group. An imprinted locus, silent on the paternal chromosome and contributing to statural growth, may be affected by the deletion.

    View details for Web of Science ID A1996VL28300006

    View details for PubMedID 8808592

  • High-density radiation hybrid map of human chromosome 18 and contig of 18p GENOMICS Giacalone, J., Li, X., Lehrach, H., FRANCKE, U. 1996; 37 (1): 9-18

    Abstract

    A high-resolution radiation hybrid map of human chromosome 18 has been developed by testing DNA samples of 92 radiation hybrids (RH) from a previously characterized chromosome 18-specific RH panel. Half of the 159 STS markers were tested on RH DNA amplified by primer extension preamplification. This map includes 20 genes and 95 polymorphic markers, most of which have heterozygosity frequencies greater than 65% and, therefore, allow integration with genetic maps. The framework map consists of 137 markers ordered with odds of 1000:1 or better and spaced on average 580 kb apart. A YAC contig map of 18p was generated independently by STS content mapping of YACs using the same primers as for RH mapping. The RH map and the contig map are concordant.

    View details for Web of Science ID A1996VL37100002

    View details for PubMedID 8921364

  • Mutant fibrillin-1 monomers lacking EGF-like domains disrupt microfibril assembly and cause severe Marfan syndrome HUMAN MOLECULAR GENETICS Liu, W. G., Qian, C. P., COMEAU, K., Brenn, T., FURTHMAYR, H., FRANCKE, U. 1996; 5 (10): 1581-1587

    Abstract

    Marfan syndrome (MFS), a heritable connective tissue disorder, is caused by mutations in the gene coding for fibrillin-1 (FBN1), an extracellular matrix protein. One of the three major categories of FBN1 mutations involves exon-skipping. To rapidly detect such mutations, we developed a long RT-PCR method. Either three segments covering the entire FBN1 coding sequence or a single 8.9 kb FBN1 coding segment were amplified from reverse-transcribed total fibroblast RNA. Restriction fragment patterns of these RT-PCR products were compared and abnormal fragments were directly sequenced. Six exon-skipping mutations were identified in a panel of 60 MFS probands. All skipped exons encode calcium binding epidermal growth factor (EGF)-like domains and maintain the reading frame. In five probands, exon-skipping was due to point mutations in splice site sequences, and one had a 6 bp deletion in a donor splice site. Pulse-chase analysis of labelled fibrillin protein revealed normal levels of synthesis but significantly reduced matrix deposition. This dominant-negative effect of the mutant monomers is considered in the light of current models of fibrillin assembly. Probands with this type of FBN1 mutation include the most severe forms of MFS, such as neonatally lethal presentations.

    View details for Web of Science ID A1996VL70200008

    View details for PubMedID 8894692

  • Chromosomal assignment of three human melanocyte-specific genes INTERNATIONAL JOURNAL OF ONCOLOGY Gratas, C., Li, X., Wang, Y. P., FRANCKE, U., Becker, D. 1996; 9 (3): 481-485

    Abstract

    To identify genes expressed in normal human melanocytes but not in malignant melanomas, we previously applied subtractive cDNA hybridization combined with PCR amplification, which led to the isolation of several human melanocyte-enriched partial cDNAs. Three of these cDNA clones were used to isolate their corresponding genomic clones. The chromosomal location of each of the three genes encoded by the individual genomic clones was determined by fluorescent in situ hybridization. The first gene mapped to human chromosome 3p14, the second gene to chromosome 19q13.1, and the third to 2p16-14. In addition, RNase A protection and in situ hybridization analyses documented expression of the gene, located on 3p14, in normal human melanocytes but not in malignant melanomas.

    View details for Web of Science ID A1996VC50000014

    View details for PubMedID 21541538

  • Dermal fibroblast culture as a model system for studies of fibrillin assembly and pathogenetic mechanisms: Defects in distinct groups of individuals with Marfan's syndrome LABORATORY INVESTIGATION Brenn, T., Aoyama, T., FRANCKE, U., FURTHMAYR, H. 1996; 75 (3): 389-402

    Abstract

    Most patients with Marfan's syndrome (> 95%) and 75% of patients with uncertain diagnosis can be classified into four groups (Aoyama et al, 1994, 1995) based on abnormal patterns of synthesis, intracellular transport, and/or matrix deposition of fibrillin-1 in fibroblast cultures. Herein we report a systematic study of fibrillin assembly in normal and Marfan's syndrome fibroblasts and correlations between pulse-chase, immunofluorescence, and immunoelectron microscopic data. Normal control fibroblasts were grown at confluent conditions from 2 to 10 days before passage and then maintained at hyperconfluent cell densities for an additional period of 1 to 6 days before assaying. Maximum deposition in the extracellular matrix of pulse-labeled fibrillin required at least 6 days of confluent and 4 to 5 days of hyperconfluent culture. This result is explained by immunofluorescence studies with fibrillin-1-specific antibodies, because 1 day after seeding cells at hyperconfluency, patches of regular immunostained structures were already present. Within these patches, fluorescence intensity and fibrillar material increased over 3 to 4 days, and after only 5 days, fibrillar networks extended throughout the culture. We propose that fibrillin-containing microfibrillar material is passaged together with the cells, newly synthesized fibrillin molecules are deposited onto preexisting microfibrillar assemblies, and several additional days of culture at high cell density are necessary for the cells to construct a sufficient microfibrillar network binding and detection of pulse-labeled fibrillin molecules in insoluble form during a 20-hour chase period. This fraction is decreased to a varying extent in fibroblast cultures of four biosynthetically distinct groups of Marfan's syndrome patients, but only Groups II and IV clearly showed reduction in immunostainable microfibrils. In long-term cultures, immunoelectron microscopy of the extracellular matrix with fibrillin antibodies also detected differences among these groups and in comparison to normal controls with respect to the arrangement of fibrillin-containing microfibrils, thickness of microfibrillar bundles, and the presence of amorphous material. The data support the idea of different pathogenetic mechanisms for each biosynthetically defined group of Marfan's syndrome, which depends on the nature of fibrillin-1 mutations.

    View details for Web of Science ID A1996VH24000011

    View details for PubMedID 8804362

  • Molecular cloning, chromosomal mapping, and characterization of the mouse UDP-galactose:ceramide galactosyltransferase gene GENOMICS Coetzee, T., Li, X., Fujita, N., Marcus, J., Suzuki, K., FRANCKE, U., Popko, B. 1996; 35 (1): 215-222

    Abstract

    UDP-galactose:ceramide galactosyltransferase (CGT) (EC 2.4.1.62) catalyzes the final step in the synthesis of galactocerebroside, a glycosphingolipid characteristically abundant in myelin. In this report, we describe the isolation of genomic clones spanning the mouse CGT gene. The mouse CGT gene consists of six exons that span a minimum of 70 kb of DNA and that encode a 541 amino acid translation product with extensive sequence similarity to the rat CGT enzyme and to UDP-glucuronosyltransferases (UGT). The 5'-untranslated region of the mouse CGT gene is encoded by a separate exon located approximately 25 kb upstream of the first protein-encoding exon. Furthermore, the genomic organization of the five coding region exons of the mouse CGT gene resembles that of the human UGT1 and rat UGT2B1 genes. Finally, analysis of somatic cell hybrids by PCR and fluorescence in situ hybridization to metaphase chromosomes has localized the mouse CGT gene to chromosome 3, bands E3-F1.

    View details for Web of Science ID A1996UV86900027

    View details for PubMedID 8661123

  • The gene for replication factor C subunit 2 (RFC2) is within the 7q11.23 Williams syndrome deletion AMERICAN JOURNAL OF HUMAN GENETICS Peoples, R., PEREZJURADO, L., Wang, Y. K., Kaplan, P., FRANCKE, U. 1996; 58 (6): 1370-1373

    View details for Web of Science ID A1996UM27100031

    View details for PubMedID 8651315

  • Wiskott-Aldrich syndrome protein, a novel effector for the GTPase CDC42Hs, is implicated in actin polymerization CELL Symons, M., Derry, J. M., Karlak, B., Jiang, S., Lemahieu, V., MCCORMICK, F., FRANCKE, U., Abo, A. 1996; 84 (5): 723-734

    Abstract

    The Rho family of GTPases control diverse biological processes, including cell morphology and mitogenesis. We have identified WASP, the protein that is defective in Wiskott-Aldrich syndrome (WAS), as a novel effector for CDC42Hs, but not for the other Rho family members, Rac and Rho. This interaction is dependent on the presence of the G protein-binding domain. Cellular expression of epitope-tagged WASP produces clusters of WASP that are highly enriched in polymerized actin. This clustering is not observed with a C-terminally deleted WASP and is inhibited by coexpression with dominant negative CDC42Hs-N17, but not with dominant negative forms of Rac or Rho. Thus, WASP provides a novel link between CDC42Hs and the actin cytoskeleton, which suggests a molecular mechanism for many of the cellular abnormalities in WAS. The WASP sequence contains two novel domains that are homologous to other proteins involved in action organization.

    View details for Web of Science ID A1996TZ99000010

    View details for PubMedID 8625410

  • The three genes of the human FCGR1 gene family encoding Fc gamma RI flank the centromere of chromosome 1 at 1p12 and 1q21 CYTOGENETICS AND CELL GENETICS Maresco, D. L., Chang, E., Theil, K. S., FRANCKE, U., Anderson, C. L. 1996; 73 (3): 157-163

    Abstract

    The high-affinity receptor for immunoglobulin G, Fc gamma RI (FCGR1), is encoded by a family of three genes within humans that share over 98% of DNA sequence homology. Efforts to define the location of the FCGR1 genes within chromosome 1 have been made to determine if they are tightly linked to the five other FCGR genes present at 1q23. Our results, obtained through both fluorescence in situ hybridization analysis of human cells and Southern analysis of cell lines containing 1p and 1q, show instead that the three genes flank the centromere of chromosome 1 at bands 1p12 and 1q21. FCGR1B was found at 1p12, whereas both FCGR1A and FCGR1C were localized to 1q21. This places the FCGR1 gene family within a large pericentric linkage group which is conserved between humans and mice. We hypothesize that the three FCGR1 genes were separated by a pericentric inversion known to have occurred on human chromosome 1, which relocated FCGR1A and FCGR1C to the long arm and left FCGR1B positioned on the short arm. We have also performed FCGR1 gene copy number experiments which indicate the existence of three FCGR1 genes within the human genome.

    View details for Web of Science ID A1996UX43200001

    View details for PubMedID 8697799

  • Assignment of the 5-hydroxytryptamine (serotonin) receptor 5A gene (HTR5A) to human chromosome band 7q36.1 CYTOGENETICS AND CELL GENETICS Schanen, N. C., Scherer, S. W., TSUI, L. C., FRANCKE, U. 1996; 72 (2-3): 187-188

    View details for Web of Science ID A1996TX31000047

    View details for PubMedID 8978771

  • Rapid evolution of human pseudoautosomal genes and their mouse homologs MAMMALIAN GENOME Ellison, J. W., Li, X., FRANCKE, U., Shapiro, L. J. 1996; 7 (1): 25-30

    Abstract

    Comparative studies of genes in the pseudoautosomal region (PAR) of human and mouse sex chromosomes have thus far been very limited. The only comparisons that can presently be made indicate that the PARs of humans and mice are not identical in terms of gene content. Here we describe additional comparative studies of human pseudoautosomal genes and their mouse homologs. Using a somatic cell hybrid mapping panel, we have assigned the mouse homolog of the human pseudoautosomal interleukin 3 receptor alpha subunit (IL3RA) gene to mouse Chromosome (Chr) 14. Attempts to clone the mouse homolog of the human pseudoautosomal adenine nucleotide translocase-3 (ANT3) gene resulted in the isolation of the murine homologs of the human ANT1 and ANT2 genes. The mouse Ant1 and Ant2 genes are very similar in sequence to their human homologs, and we have mapped them to mouse Chromosomes (Chrs) (8 and X respectively) that exhibit conserved synteny with the chromosomes on which the human genes are located. In contrast, the homolog of ANT3 appears to be either very divergent or absent from the mouse genome. Southern blot analysis of DNA from a variety of mammalian species shows restricted conservation of human pseudoautosomal genes, a trend that also applies to the two cloned mouse homologs of these genes and to neighboring human genes in distal Xp22.3. Our observations combined with those of other workers lead us to propose a model for the evolution of the PAR that includes both rapid sequence evolution and the incremental reduction in size of the region during mammalian evolution.

    View details for Web of Science ID A1996TQ96700007

    View details for PubMedID 8903724

  • The IPW gene is imprinted and is not expressed in the Prader-Willi syndrome. Acta geneticae medicae et gemellologiae Wevrick, R., Kerns, J. A., FRANCKE, U. 1996; 45 (1-2): 191-197

    View details for PubMedID 8872030

  • RBM3, A NOVEL HUMAN GENE IN XP11.23 WITH A PUTATIVE RNA-BINDING DOMAIN HUMAN MOLECULAR GENETICS Derry, J. M., Kerns, J. A., FRANCKE, U. 1995; 4 (12): 2307-2311

    Abstract

    Using cDNA selection with a YAC from the Xp11.2 region, we have identified a novel gene (RBM3) that encodes a polypeptide with high sequence similarity to a group of proteins that bind to RNA. On a YAC contig map, RBM3 is located between OATL1 and GATA1/TFE3 in sub-band Xp11.23, and gives rise to alternatively spliced transcripts in a variety of human tissues. The longest open reading frame encodes a 157 amino acid protein with a predicted molecular weight of 17 kDa. Its putative RNA-binding domain most closely resembles that of two previously characterized human RNA-binding proteins, YRRM, the gene for which has been implicated in azoospermia, and hnRNP G, a glycoprotein, also identified as an auto-antigen. The homology of RMB3 in both sequence and size to an RNA binding protein from maize, AAIP , suggests that it functions in a fundamental pathway conserved from plants to mammals.

    View details for Web of Science ID A1995TK36300017

    View details for PubMedID 8634703

  • CLONING AND CHARACTERIZATION OF A NOVEL ZINC-FINGER GENE IN XP11.2 GENOMICS Derry, J. M., Jess, U., FRANCKE, U. 1995; 30 (2): 361-365

    Abstract

    During a systematic search for open reading frames in chromosome band Xp11.2, a novel gene (ZNF157) that encodes a putative 506-amino-acid protein with the sequence characteristics of a zinc-finger-containing transcription factor was isolated. ZNF157 is encoded by four exons distributed over > 20 kb of genomic DNA. The second and third exons contain sequences similar to those of the previously described KRAB-A and KRAB-B domains, motifs that have been shown to mediate transcriptional repression in other members of the protein family. A fourth exon contains 12 zinc finger DNA binding motifs and finger linking regions characteristic of ZNF proteins of the Krüppel family. ZNF157 maps to the telomeric end of a cluster of ZNF genes that includes ZNF21, ZNF41, and ZNF81.

    View details for Web of Science ID A1995TG44700031

    View details for PubMedID 8586441

  • THE WISKOTT-ALDRICH SYNDROME AND X-LINKED CONGENITAL THROMBOCYTOPENIA ARE CAUSED BY MUTATIONS OF THE SAME GENE BLOOD Zhu, Q. L., Zhang, M., BLAESE, R. M., Derry, J. M., Junker, A., FRANCKE, U., Chen, S. H., Ochs, H. D. 1995; 86 (10): 3797-3804

    Abstract

    The Wiskott-Aldrich syndrome (WAS) is an X-linked recessive disorder characterized by thrombocytopenia, small platelets, eczema, recurrent infections, and immunodeficiency. Besides the classic WAS phenotype, there is a group of patients with congenital X-linked thrombocytopenia (XLT) who have small platelets but only transient eczema, if any, and minimal immune deficiency. Because the gene responsible for WAS has been sequenced, it was possible to correlate the WAS phenotypes with WAS gene mutations. Using a fingerprinting screening technique, we determined the approximate location of the mutation in 13 unrelated WAS patients with mild to severe clinical symptoms. Direct sequence analysis of cDNA and genomic DNA obtained from patient-derived cell lines showed 12 unique mutations distributed throughout the WAS gene, including insertions, deletions, and point mutations resulting in amino acid substitutions, termination, exon skipping, or splicing defects. Of 4 unrelated patients with the XLT phenotype, 3 had missense mutations affecting exon 2 and 1 had a splice-site mutation affecting exon 9. Patients with classic WAS had more complex mutations, resulting in termination codons, frameshift, and early termination. These findings provide direct evidence that XLT and WAS are caused by mutations of the same gene and suggest that severe clinical phenotypes are associated with complex mutations.

    View details for Web of Science ID A1995TE58500021

    View details for PubMedID 7579347

  • GENETIC-MAPPING OF THE HUMAN AMPHIPHYSIN GENE (AMPH) AT 7P14-P13 EXCLUDES ITS INVOLVEMENT IN RETINITIS-PIGMENTOSA-9 OR DOMINANT CYSTOID MACULAR DYSTROPHY AMERICAN JOURNAL OF HUMAN GENETICS Yamamoto, R., Li, X., FRANCKE, U., Kilimann, M. W. 1995; 57 (4): 970-972

    View details for Web of Science ID A1995RV79300033

    View details for PubMedID 7573063

  • THE MOUSE HOMOLOG OF THE WISKOTT-ALDRICH SYNDROME PROTEIN (WASP) GENE IS HIGHLY CONSERVED AND MAPS NEAR THE SCURFY (SF) MUTATION ON THE X-CHROMOSOME GENOMICS Derry, J. M., Wiedemann, P., Blair, P., Wang, Y., Kerns, J. A., Lemahieu, V., Godfrey, V. L., Wilkinson, J. E., FRANCKE, U. 1995; 29 (2): 471-477

    Abstract

    The mouse WASP gene, the homolog of the gene mutated in Wiskott-Aldrich syndrome, has been isolated and sequenced. the predicted amino acid sequence is 86% identical to the human WASP sequence. A distinct feature of the mouse gene is an expanded polymorphic GGA trinucleotide repeat that codes for polyglycine and varies from 15 to 17 triplets in different Mus musculus strains. The genomic structure of the mouse WASP gene is expressed as an approximately 2.4-kb mRNA in thymus and spleen. Chromosomal mapping in an interspecific M. Musculus/M. spretus backcross placed the Wasp locus near the centromere of the mouse X chromosome, inseparable from Gata1, Tcfe3, and scurfy (sf). This localization makes Wasp a candidate for involvement in scurfy, a T cell-mediated fatal lymphoreticular disease of mice that has previously been proposed as a mouse homolog of Wiskott-Aldrich syndrome. Northern analysis of sf tissue samples indicated the presence of WASP mRNA in liver and skin, presumably as a consequence of lymphocytic infiltration, but non abnormalities in the amount or size of mRNA present.

    View details for Web of Science ID A1995RY05500021

    View details for PubMedID 8666397

  • ORGANIZATION, REGULATORY SEQUENCES, AND ALTERNATIVELY SPLICED TRANSCRIPTS OF THE MUCOSAL ADDRESSIN CELL-ADHESION MOLECULE-1 (MADCAM-1) GENE JOURNAL OF IMMUNOLOGY SAMPAIO, S. O., Li, X., Takeuchi, M., Mei, C., FRANCKE, U., BUTCHER, E. C., Briskin, M. J. 1995; 155 (5): 2477-2486

    Abstract

    The mucosal addressin cell adhesion molecule-1 (MAdCAM-1) is expressed selectively at venular sites of lymphocyte extravasation into mucosal lymphoid tissues and lamina propria, where it directs local lymphocyte trafficking. MAdCAM-1 is a multifunctional type l transmembrane adhesion molecule comprising two distal lg domains involved in alpha 4 beta 7 integrin binding, a mucin-like region able to display L-selectin-binding carbohydrates, and a membrane-proximal lg domain homologous to lgA. We show in this work that the MAdCAM-1 gene is located on chromosome 10 and contains five exons. The signal peptide and each one of the three lg domains are encoded by a distinct exon, whereas the transmembrane, cytoplasmic tail, and 3'-untranslated region of MAdCAM-1 are combined on a single exon. The mucin-like region and the third lg domain are encoded together on exon 4. An alternatively spliced MAdCAM-1 mRNA is identified that lacks the mucin/lgA-homologous exon 4-encoded sequences. This short variant of MAdCAM-1 may be specialized to support alpha 4 beta 7-dependent adhesion strengthening, independent of carbohydrate-presenting function. Sequences 5' of the transcription start site include tandem nuclear factor-kappa B sites; AP-1, AP-2, and signal peptide-1 binding sites; and an estrogen response element. Our findings reinforce the correspondence between the multidomain structure and versatile functions of this vascular addressin, and suggest an additional level of regulation of carbohydrate-presenting capability, and thus of its importance in lectin-mediated vs alpha 4 beta 7-dependent adhesive events in lymphocyte trafficking.

    View details for Web of Science ID A1995RP74300021

    View details for PubMedID 7650378

  • FIBRILLIN ABNORMALITIES AND PROGNOSIS IN MARFAN-SYNDROME AND RELATED DISORDERS AMERICAN JOURNAL OF MEDICAL GENETICS Aoyama, T., FRANCKE, U., Gasner, C., FURTHMAYR, H. 1995; 58 (2): 169-176

    Abstract

    Marfan syndrome (MFS), a multisystem autosomal-dominant disorder, is characterized by mutations of the fibrillin-1 (FBN1) gene and by abnormal patterns of synthesis, secretion, and matrix deposition of the fibrillin protein. To determine the sensitivity and specificity of fibrillin protein abnormalities in the diagnosis of MFS, we studied dermal fibroblasts from 57 patients with classical MFS, 15 with equivocal MFS, 8 with single-organ manifestations, and 16 with other connective tissue disorders including homocystinuria and Ehlers-Danlos syndrome. Abnormal fibrillin metabolism was identified in 70 samples that were classified into four different groups based on quantitation of fibrillin synthesis and matrix deposition. Significant correlations were found for phenotypic features including arachnodactyly, striae distensae, cardiovascular manifestations, and fibrillin groups II and IV, which included 70% of the MFS patients. In addition, these two groups were associated with shortened "event-free" survival and more severe cardiovascular complications than groups I and III. The latter included most of the equivocal MFS/single manifestation patients with fibrillin abnormalities. Our results indicate that fibrillin defects at the protein level per se are not specific for MFS, but that the drastically reduced fibrillin deposition, caused by a dominant-negative effect of abnormal fibrillin molecules in individuals defined as groups II and IV, is of prognostic and possibly diagnostic significance.

    View details for Web of Science ID A1995RP83600015

    View details for PubMedID 8533811

  • NFATC3, A LYMPHOID-SPECIFIC NFATC FAMILY MEMBER THAT IS CALCIUM-REGULATED AND EXHIBITS DISTINCT DNA FINDING SPECIFICITY JOURNAL OF BIOLOGICAL CHEMISTRY Ho, S. N., Thomas, D. J., Timmerman, L. A., Li, X., FRANCKE, U., Crabtree, G. R. 1995; 270 (34): 19898-19907

    Abstract

    Signals transduced by the T cell antigen receptor (TCR) regulate developmental transitions in the thymus and also mediate the immunologic activation of mature, peripheral T cells. In both cases TCR stimulation leads to the assembly of the NFAT transcription complex as a result of the calcium-dependent nuclear translocation of cytosolic subunits, NFATc, and the Ras/protein kinase C-dependent induction of a nuclear subunit, NFATn. To further understand the diverse roles of antigen receptor signaling throughout T cell development, we have identified a new NFATc family member, NFATc3, that is expressed at highest levels in the thymus. NFATc3 is the product of a gene on murine chromosome 8 that is not linked to the other NFATc genes. NFATc3, like other NFATc family members, contains a conserved rel similarity domain, and also defines a region conserved among NFATc family members, the SP repeat region, characterized by the repeated motif SPxxSPxxSPrxsxx (D/E)(D/E)swl. NFATc3 activates NFAT site-dependent transcription when overexpressed, yet exhibits a pattern of DNA site specificity distinct from other NFATc proteins. Additionally, thymic NFATc3 undergoes modifications in response to agents that mimic T cell receptor signaling, including a decrease in apparent molecular mass upon elevation of intracellular calcium that is inhibited by the immunosuppressant FK506. Given the preferential expression of NFATc3 in the thymus, NFATc family members may regulate distinct subsets of genes during T cell development.

    View details for PubMedID 7650004

  • MOLECULAR-CLONING OF THE HUMAN HOMOLOG OF A STRIATUM-ENRICHED PHOSPHATASE (STEP) GENE AND CHROMOSOMAL MAPPING OF THE HUMAN AND MURINE LOCI GENOMICS Li, X., Luna, J., Lombroso, P. J., FRANCKE, U. 1995; 28 (3): 442-449

    Abstract

    A gene for a protein tyrosine phosphatase (PTPase) was isolated from a human fetal brain cDNA library by PCR amplification. Sequence analysis revealed that the PTPase has a single phosphatase catalytic domain located at the C-terminus that includes the highly conserved amino acid domain [I/V]HCXAGXXR[S/T]GX[F/Y] found in all tyrosine phosphatases. Two proline-rich regions located at the N-terminus may contain putative Src homology domain 3 (SH3) binding motifs. Comparison of the PTPase with a previously cloned striatum enriched phosphatase (STEP) from rat and from mouse exhibited a high degree of identity (approximately 85-90%) at both the nucleotide and the amino acid levels, indicating that the human PTPase is the homolog of the rat and murine STEP gene. By using a combination of somatic cell hybrid analysis and fluorescence in situ hybridization, we have mapped the human STEP locus to chromosome 11p15.2-p15.1 and the murine STEP gene to chromosome 7B3-B5. These are two regions of known conserved synteny, providing further evidence that the human STEP is a true homolog of the murine STEP gene. Candidate disease genes in the vicinity include Usher syndrome type 1C in human and a mouse mutant locus, twister (twt).

    View details for Web of Science ID A1995RQ98900011

    View details for PubMedID 7490079

  • HUMAN PEROXISOMAL TARGETING SIGNAL-1 RECEPTOR RESTORES PEROXISOMAL PROTEIN IMPORT IN CELLS FROM PATIENTS WITH FATAL PEROXISOMAL DISORDERS JOURNAL OF CELL BIOLOGY Wiemer, E. A., Nuttley, W. M., Bertolaet, B. L., Li, X., FRANCKE, U., Wheelock, M. J., ANNE, U. K., Johnson, K. R., Subramani, S. 1995; 130 (1): 51-65

    Abstract

    Two peroxisomal targeting signals, PTS1 and PTS2, are involved in the import of proteins into the peroxisome matrix. Human patients with fatal generalized peroxisomal deficiency disorders fall into at least nine genetic complementation groups. Cells from many of these patients are deficient in the import of PTS1-containing proteins, but the causes of the protein-import defect in these patients are unknown. We have cloned and sequenced the human cDNA homologue (PTS1R) of the Pichia pastoris PAS8 gene, the PTS1 receptor (McCollum, D., E. Monosov, and S. Subramani. 1993. J. Cell Biol. 121:761-774). The PTS1R mRNA is expressed in all human tissues examined. Antibodies to the human PTS1R recognize this protein in human, monkey, rat, and hamster cells. The protein is localized mainly in the cytosol but is also found to be associated with peroxisomes. Part of the peroxisomal PTS1R protein is tightly bound to the peroxisomal membrane. Antibodies to PTS1R inhibit peroxisomal protein-import of PTS1-containing proteins in a permeabilized CHO cell system. In vitro-translated PTS1R protein specifically binds a serine-lysine-leucine-peptide. A PAS8-PTS1R fusion protein complements the P. pastoris pas8 mutant. The PTS1R cDNA also complements the PTS1 protein-import defect in skin fibroblasts from patients--belonging to complementation group two--diagnosed as having neonatal adrenoleukodystrophy or Zellweger syndrome. The PTS1R gene has been localized to a chromosomal location where no other peroxisomal disorder genes are known to map. Our findings represent the only case in which the molecular basis of the protein-import deficiency in human peroxisomal disorders is understood.

    View details for Web of Science ID A1995RF98600005

    View details for PubMedID 7790377

  • SEVERE GROWTH-HORMONE INSENSITIVITY (LARON SYNDROME) DUE TO NONSENSE MUTATION OF THE GH RECEPTOR IN BROTHERS FROM RUSSIA JOURNAL OF PEDIATRIC ENDOCRINOLOGY & METABOLISM Rosenbloom, A. L., Berg, M. A., Kasatkina, E. P., VOLKOVA, T. N., SKOROBOGATOVA, V. F., SOKOLOVSKAYA, V. N., FRANCKE, U. 1995; 8 (3): 159-165

    Abstract

    Primary GH insensitivity (Laron syndrome) due to GH receptor deficiency (GHRD) is an autosomal recessive condition characterized by severe growth failure. Diverse alterations in the GHR gene have been reported in affected individuals. We report here the first family with GHRD from Russia, with two affected siblings and consanguineous parents. Analysis of blood spot DNA by polymerase chain reaction (PCR), denaturing gradiant gel electrophoresis, and nucleotide sequencing indicated that these siblings are homozygous for a nonsense mutation, R43X, in the GHR gene. The R43X mutation, which changes an arginine codon to a translational stop codon, occurs at a CpG dinucleotide mutational hotspot and has previously been identified in affected individuals of Mediterranean and Ecuadorian origin.

    View details for Web of Science ID A1995RU79100002

    View details for PubMedID 8521189

  • WASP GENE-MUTATIONS IN WISKOTT-ALDRICH SYNDROME AND X-LINKED THROMBOCYTOPENIA HUMAN MOLECULAR GENETICS Derry, J. M., Kerns, J. A., Weinberg, K. I., Ochs, H. D., Volpini, V., Estivill, X., Walker, A. P., FRANCKE, U. 1995; 4 (7): 1127-1135

    Abstract

    The WASP gene has been recently cloned from Xp11.23 and shown to be mutated in three patients with the Wiskott-Aldrich syndrome (WAS). We have developed a screening protocol for identifying WASP gene alterations in genomic DNA and have identified a spectrum of novel mutations in 12 additional unrelated families. These missense, nonsense and frameshift mutations involve eight of the 12 exons of the gene. Two mutations creating premature termination codons were associated with lack of detectable mRNA on Northern blots. Four amino acid substitutions, Leu27Phe, Thr48Ile, Val75Met and Arg477Lys, were found in patients with congenital thrombocytopenia and no clinically evident immune defect indicating that the WASP gene is the site for mutations in X-linked thrombocytopenia as well as in WAS. A T-cell line from a WAS patient contained two independent DNA alterations, a constitutional frameshift mutation, also present in peripheral blood leukocytes from the patient, and compensatory splice site mutation unique to the cell line. The distribution of eight missense mutations provides valuable information on amino acids which are essential for normal protein function, and suggests that sites in the first two exons are hot-spots for mutation.

    View details for Web of Science ID A1995RG49900002

    View details for PubMedID 8528199

  • CLONING, MAPPING, AND CHARACTERIZATION OF ACTIVATED LEUKOCYTE-CELL ADHESION MOLECULE (ALCAM), A CD6 LIGAND JOURNAL OF EXPERIMENTAL MEDICINE Bowen, M. A., Patel, D. D., Li, X., MODRELL, B., Malacko, A. R., Wang, W. C., MARQUARDT, H., Neubauer, M., Pesando, J. M., FRANCKE, U., HAYNES, B. F., Aruffo, A. 1995; 181 (6): 2213-2220

    Abstract

    Antibody-blocking studies have demonstrated the role of CD6 in thymocyte-thymic epithelial (TE) cell adhesion. Here we report that CD6 expressed by COS cells mediates adhesion to TE cells and that this interaction is specifically blocked with an anti-CD6 monoclonal antibody (mAb) or with a mAb (J4-81) that recognized a TE cell antigen. We isolated and expressed a cDNA clone encoding this antigen and show that COS cells transfected with this cDNA bind a CD6 immunoglobulin fusion protein (CD6-Rg). This antigen, which we named ALCAM (activated leukocyte-cell adhesion molecule) because of its expression on activated leukocytes, appears to be the human homologue of the chicken neural adhesion molecule BEN/SC-1/DM-GRASP. The gene was mapped to human chromosome 3q13.1-q13.2 by fluorescence in situ hybridization of cDNA probes to metaphase chromosomes. We prepared an ALCAM-Rg fusion protein and showed that it binds to COS cell transfectants expressing CD6, demonstrating that ALCAM is a CD6 ligand. The observations that ALCAM is also expressed by activated leukocytes and that both ALCAM and CD6 are expressed in the brain suggest that ALCAM-CD6 interactions may play a role in the binding of T and B cells to activated leukocytes, as well as in interactions between cells of the nervous system.

    View details for Web of Science ID A1995RA60500029

    View details for PubMedID 7760007

  • A GLY1127SER MUTATION IN AN EGF-LIKE DOMAIN OF THE FIBRILLIN-1 GENE IS A RISK FACTOR FOR ASCENDING AORTIC-ANEURYSM AND DISSECTION AMERICAN JOURNAL OF HUMAN GENETICS FRANCKE, U., Berg, M. A., Tynan, K., Brenn, T., Liu, W. G., Aoyama, T., Gasner, C., Miller, D. C., FURTHMAYR, H. 1995; 56 (6): 1287-1296

    Abstract

    Ascending aortic disease, ranging from mild aortic root enlargement to aneurysm and/or dissection, has been identified in 10 individuals of a kindred, none of whom had classical Marfan syndrome (MFS). Single-strand conformation analysis of the entire fibrillin-1 (FBN1) cDNA of an affected family member revealed a G-to-A transition at nucleotide 3379, predicting a Gly1127Ser substitution. The glycine in this position is highly conserved in EGF-like domains of FBN1 and other proteins. This mutation was present in 9 of 10 affected family members and in 1 young unaffected member but was not found in other unaffected members, in 168 chromosomes from normal controls, and in 188 chromosomes from other individuals with MFS or related phenotypes. FBN1 intragenic marker haplotypes ruled out the possibility that the other allele played a significant role in modulating the phenotype in this family. Pulse-chase studies revealed normal fibrillin synthesis but reduced fibrillin deposition into the extracellular matrix in cultured fibroblasts from a Gly1127Ser carrier. We postulate that the Gly1127Ser FBN1 mutation is responsible for reduced matrix deposition. We suggest that mutations such as this one may disrupt EGF-like domain folding less drastically than do substitutions of cysteine or of other amino acids important for calcium-binding that cause classical MFS. The Gly1127Ser mutation, therefore, produces a mild form of autosomal dominantly inherited weakness of elastic tissue, which predisposes to ascending aortic aneurysm and dissection later in life.

    View details for PubMedID 7762551

  • CONSTITUTIONAL TRANSLOCATION T(1-17)(P36.31-P36.13-Q11.2Q12.1) IN A NEUROBLASTOMA PATIENT. ESTABLISHMENT OF SOMATIC-CELL HYBRIDS AND IDENTIFICATION OF PND/A12M2 ON CHROMOSOME-1 AND NF1/SCYA7 ON CHROMOSOME-17 AS BREAKPOINT FLANKING SINGLE-COPY MARKERS ONCOGENE Laureys, G., Speleman, F., Versteeg, R., VANDERDRIFT, P., Chan, A., Leroy, J., FRANCKE, U., Opdenakker, G., VANROY, N. 1995; 10 (6): 1087-1093

    Abstract

    Cytogenetic and molecular studies in neuroblastoma suggest the presence of a tumor suppressor gene at the distal band p36 of human chromosome 1. We described a constitutional translocation t(1;17)(p36;q12-q21), involving the critical region 1p36, in a patient with neuroblastoma, and hypothesized that the translocation predisposed the patient to tumor development. Here we report the molecular delineation of the translocation breakpoints. Somatic cell hybrids were generated by fusion of the patient's fibroblasts with the thymidine kinase deficient hamster cell line, a3. In hybrid cell lines which retained the human derivative chromosomes, the position of chromosome 1p and 17q DNA probes respective to the translocation breakpoints was determined by fluorescence in situ hybridization and Southern blot analysis. The chromosome 1p breakpoint was localized within a repetitive region encoding t-RNA genes, with 12A-2 (PND) as most distal and pHE2.6 (A12M2) as most proximal single-copy breakpoint flanking markers. For the chromosome 17 breakpoint, the proximal and distal flanking markers were identified as 7G4 (NF1) and cMCP-3 (SCYA7), respectively. In this study, cMCP-3 (SCYA7), encoding the human monocyte chemotactic protein-3, was mapped between NF1 and ERBB2. As a pivotal step towards breakpoint cloning, at present these flanking markers optimally delineate the breakpoint regions of both chromosomes 1 and 17 at the molecular level.

    View details for Web of Science ID A1995QN35300008

    View details for PubMedID 7700633

  • THE QUESTION OF HETEROGENEITY IN MARFAN-SYNDROME NATURE GENETICS Dietz, H., FRANCKE, U., FURTHMAYR, H., Francomano, C., Depaepe, A., Devereux, R., Ramirez, F., Pyeritz, R. 1995; 9 (3): 228-229

    View details for Web of Science ID A1995QL23200007

    View details for PubMedID 7773282

  • CHARACTERIZATION OF THE HUMAN GENE FOR MICROFIBRIL-ASSOCIATED GLYCOPROTEIN (MFAP2), ASSIGNMENT TO CHROMOSOME 1P36.1-P35, AND LINKAGE TO D1S170 GENOMICS Faraco, J., Bashir, M., Rosenbloom, J., FRANCKE, U. 1995; 25 (3): 630-637

    Abstract

    Microfibril-associated glycoprotein, MAGP (gene symbol MFAP2), is a component of connective tissue microfibrils and a candidate for involvement in the etiology of inherited connective tissue diseases. We have cloned a human MAGP cDNA that is highly homologous to the previously characterized bovine and murine genes. Like the bovine and murine loci, the human gene has eight coding exons, but it contains two alternatively used 5' untranslated exons, whereas only one untranslated exon was described in the bovine and murine Magp genes. By using rodent x human somatic cell hybrid panels and fluorescence chromosomal in situ hybridization, we have assigned the locus to human chromosome 1p36.1-p35. An insertion/deletion polymorphism has been identified within intron 7. Linkage analysis between this polymorphism and markers on distal chromosome 1 revealed that MAGP is tightly linked to the anonymous marker D1S170. Physical mapping revealed a distance of < 100 kb between the two markers. This information can be used to screen for linkage in families with microfibrillar abnormalities that are not linked to the fibrillin genes on chromosomes 15 or 5.

    View details for Web of Science ID A1995QL41600004

    View details for PubMedID 7759096

  • PRIMARY STRUCTURE OF HUMAN AMPHIPHYSIN, THE DOMINANT - AUTOANTIGEN OF PARANEOPLASTIC STIFF-MAN SYNDROME, AND MAPPING OF ITS GENE (AMPH) TO CHROMOSOME 7P13-P14 HUMAN MOLECULAR GENETICS Yamamoto, R., Li, X., Winter, S., FRANCKE, U., Kilimann, M. W. 1995; 4 (2): 265-268

    Abstract

    Amphiphysin is a protein peripherally associated with synaptic vesicles. It is expressed in many neurons, certain endocrine cell types, and spermatocytes. Autoantibodies against amphiphysin occur in patients afflicted with a rare neurologic autoimmune disease, paraneoplastic Stiff-Man syndrome. To provide a basis for the understanding of anti-amphiphysin autoimmunity, we have cloned cDNAs and determined the primary structure of human amphiphysin. Comparison with chicken amphiphysin defines domains of low and high amino acid sequence conservation. As a candidate for heritable disorders of the nervous system, endocrine tissues or male fertility, the human amphiphysin gene was mapped to chromosome 7, region p13-p14.

    View details for Web of Science ID A1995QH63400018

    View details for PubMedID 7757077

  • ASSIGNMENT OF THE GENE SLC1A2 CODING FOR THE HUMAN GLUTAMATE TRANSPORTER EAAT2 TO HUMAN-CHROMOSOME-11 BANDS P13-P12 CYTOGENETICS AND CELL GENETICS Li, X., FRANCKE, U. 1995; 71 (3): 212-213

    View details for Web of Science ID A1995TD38500002

    View details for PubMedID 7587378

  • Clinical and molecular cytogenetics and gene mapping: principles and techniques. Southeast Asian journal of tropical medicine and public health FRANCKE, U. 1995; 26: 34-43

    Abstract

    This article reviews the history of human cytogenetics with respect to technical advances from chromosome banding to molecular cytogenetics. Technologies such as in situ hybridization, chromosome painting, comparative genomic hybridization and interphase cytogenetics and their applications are discussed. The assignments of genes to chromosome regions by somatic cell genetics is illustrated by molecular analyses of somatic cell hybrid panels. The generation of complete physical maps of human chromosomes, by radiation hybrid mapping of sequence-tagged sites and establishment of chromosome-specific yeast artificial chromosome (YAC) banks and clone overlaps (contigs), is exemplified by studies of chromosome 18. The last section outlines the recent and future advances in clinical cytogenetics made possible by progress in molecular genetics.

    View details for PubMedID 8629138

  • CLONING AND REGIONAL ASSIGNMENT OF THE HUMAN MYOSIN HEAVY-CHAIN-12 (MYH12) GENE TO CHROMOSOME BAND-15Q21 CYTOGENETICS AND CELL GENETICS Moore, K. J., Testa, J. R., FRANCKE, U., MILATOVICH, A., Copeland, N. G., Jenkins, N. A. 1995; 69 (1-2): 53-58

    Abstract

    Sequences encoding 1,235 bp of the human myosin heavy chain 12 (MYH12) gene have been cloned from a human brain cDNA library by PCR amplification. The human sequence is 95.8% identical to the mouse sequence at the amino acid level, indicating that the MYH12 gene has been evolutionarily well conserved. Somatic cell hybrid analysis and in situ hybridization place the MYH12 gene on human chromosome 15, at band q21, and extend distally the known region of chromosome 15 linkage homology on mouse chromosome 9.

    View details for Web of Science ID A1995QF04500013

    View details for PubMedID 7835087

  • MAPPING OF HUMAN AND MURINE GENES FOR LATENT TGF-BETA BINDING PROTEIN-2 (LTBP2) MAMMALIAN GENOME Li, X., Yin, W., PEREZJURADO, L., Bonadio, J., FRANCKE, U. 1995; 6 (1): 42-45

    Abstract

    A novel gene, isolated because of structural similarities to fibrillin, was called LTBP2 when its 4.6-kb transcript was found to encode a protein sequence related to the latent TGF-beta binding protein (LTBP1), which is encoded on human chromosome (Chr) 2, region p12-q22. We have assigned the human and murine LTBP2 loci to regions of conserved synteny on human Chr 11 and mouse Chr 19. By PCR analysis of somatic cell hybrid DNA and fluorescence in situ hybridization, LTBP2 was mapped to human Chr band 11q12 and Ltbp2 to mouse Chr band 19B. Differences between inbred strains were discovered by single-strand conformation analysis of PCR products from the 3' untranslated region. Analysis of BXD and AKXL recombinant inbred strains have placed Ltbp2 between D19Rp19 and Ly10 on proximal mouse Chr 19.

    View details for Web of Science ID A1995QB67100008

    View details for PubMedID 7719025

  • CLONING AND CHROMOSOMAL LOCALIZATION OF THE HUMAN AND MURINE GENES FOR THE T-CELL TRANSCRIPTION FACTORS NFATC AND NFATP CYTOGENETICS AND CELL GENETICS Li, X., Ho, S. N., Luna, J., Giacalone, J., Thomas, D. J., Timmerman, L. A., Crabtree, G. R., FRANCKE, U. 1995; 68 (3-4): 185-191

    Abstract

    The nuclear factor of activated T cells (NFAT) is a transcription factor complex involved in the activation of cytokines and cell surface molecules associated with coordinating the actions of different cells required for an immune response. Two different genes have recently been cloned that encode proteins capable of functioning as the pre-existing (p) and cytosolic (c) component of the NFAT transcription complex, NFATc of human and NFATp of murine origin (Northrop et al., 1994; McCaffrey et al., 1993b). We report here the partial cDNA cloning of the murine homolog of NFATc and the human homolog of NFATp, and the chromosomal localization of both genes in both species to conserved syntenic regions. Through the use of mapping panels of human x Chinese hamster and mouse x rodent cells hybrids, the NFATc genes were mapped to human and mouse chromosomes 18. By analyzing a chromosome 18 radiation hybrid panel, the human NFATc gene was localized to the q terminus, closely linked to STS marker D18S497. The murine Nfatc gene was sublocalized to chromosome band 18E4 by FISH. The NFATp genes were mapped by somatic cell hybrid analysis to human chromosome 20 and mouse chromosome 2. Human NFATp was assigned to chromosome region 20q13.2-->q13.3 by FISH. Based on the conserved syntenic region on human chromosome 20 and mouse chromosome 2, murine Nfatp is predicted to reside in the vicinity of a mutant locus wasted. Homozygous wst/wst mice display a phenotype reminiscent of severe combined immune deficiency or ataxia telangiectasia, disorders that could therefore be considered candidates for NFATp mutations.

    View details for PubMedID 7842733

  • HEMIZYGOSITY AT THE INSULIN-LIKE GROWTH-FACTOR-I RECEPTOR (IGF1R) LOCUS AND GROWTH FAILURE IN THE RING CHROMOSOME-15 SYNDROME CYTOGENETICS AND CELL GENETICS Peoples, R., MILATOVICH, A., FRANCKE, U. 1995; 70 (3-4): 228-234

    Abstract

    The ring chromosome 15 syndrome is characterized by mild-to-severe growth failure. We evaluated the status of the insulin-like growth factor I receptor (IGF1R) gene, which had previously been assigned to band 15q26 in several patients with de novo ring 15 chromosomes, to investigate a possible correlation between disruption or loss of the IGF1R gene with the severe growth failure phenotype. The presence or absence of the IGF1R gene on the ring 15 chromosomes of five patients was ascertained by in situ hybridization and gene-dosage (Southern) blotting. The location of the breakpoints was determined by typing polymorphic markers from the distal end of the long arm of chromosome 15 in both the probands and their parents. Deletion mapping determined that all breakpoints were distal to D15S100 and that the IGF1R gene is located between D15S107 and D15S87. Three patients who had suffered severe growth failure in early childhood were hemizygous at the IGF1R locus, while one patient with borderline short stature had two copies of the IGF1R gene. The correlation between IGF1R gene dosage and growth retardation demonstrated here in our ring chromosome 15 patients suggests a possible role for heterozygous IGF1R gene mutations or deletions in other cases of unexplained growth failure.

    View details for Web of Science ID A1995RG53300067

    View details for PubMedID 7789178

  • CHARACTERIZATION OF THE CHROMOSOME BREAKPOINTS IN A PATIENT WITH A CONSTITUTIONAL TRANSLOCATION T(1-17)(P36.31-P36.13-Q11.2-Q12) AND NEUROBLASTOMA EUROPEAN JOURNAL OF CANCER Laureys, G., Versteeg, R., Speleman, F., VANDERDRIFT, P., FRANCKE, U., Opdenakker, G., VANROY, N. 1995; 31A (4): 523-526

    Abstract

    Cytogenetic and molecular studies in neuroblastoma suggest the presence of a tumour suppressor gene at the distal chromosome band 1p36. Previously, we hypothesised that a constitutional translocation involving the region 1p36 [t(1;17)(p36;q12-q21)] in a patient with neuroblastoma predisposed him to tumour development. Here we report the molecular delineation of the translocation breakpoints. Somatic cell hybrids containing the derivative chromosomes were used to determine the position of chromosome 1p and 17q DNA probes respective to the breakpoints using fluorescence in situ hybridisation. The 1p breakpoint was localised between the PND and D1S56 loci. The chromosome 17q breakpoint is flanked by NF1 and SCYA7, as proximal and distal marker, respectively. We redefined the translocation as t(1;17)(p36.31-13;q11.2-q12). The identification of flanking markers of the breakpoints is a prerequisite for breakpoint cloning and identification of a putative neuroblastoma suppressor gene.

    View details for Web of Science ID A1995QY18000021

    View details for PubMedID 7576958

  • MAPPING OF SYNAPSIN-II (SYN2) GENES TO HUMAN-CHROMOSOME-3P AND MOUSE CHROMOSOME-6 BAND-F CYTOGENETICS AND CELL GENETICS Li, X., Rosahl, T. W., Sudhof, T. C., FRANCKE, U. 1995; 71 (3): 301-305

    Abstract

    Synapsins are neuron-specific phosphoproteins of small synaptic vesicles encoded by two different genes. While the gene for synapsin I (SYN1) is on the X chromosome, we have now assigned the human and mouse synapsin II (SYN2) genes to autosomes. By using PCR primers derived from rat synapsin II cDNA sequences we were able to amplify homologous sequences of the 3'-untranslated regions and to localize the human SYN2 gene to 3p and the mouse Syn2 gene to mouse chromosome 6 by single strand conformation analysis of PCR products from panels of somatic hybrid cell lines. The mouse gene was further mapped by FISH to chromosome 6 band F in a region of known conserved synteny with human 3p. Genotyping of a M. musculus x M. spretus backcross panel placed Syn2 close to a cluster of previously mapped loci on chromosome 6 in an interval between interleukin 5 receptor alpha (Il5ra) and hematopoietic cell phosphatase 1C (Hcph). Both physical and genetic mapping data indicate that Syn2 is near two mutant loci defined by neuromuscular disorders, opisthotonus (opt) and deaf waddler (dfw).

    View details for Web of Science ID A1995TD38500023

    View details for PubMedID 7587399

  • THE HUMAN CALCITONIN RECEPTOR GENE (CALCR) AT 7Q21.3 IS OUTSIDE THE DELETION ASSOCIATED WITH THE WILLIAMS-SYNDROME CYTOGENETICS AND CELL GENETICS Jurado, L. A., Li, X., FRANCKE, U. 1995; 70 (3-4): 246-249

    Abstract

    The human calcitonin receptor (CTR) is a transmembrane peptide with dual action as a receptor for the hormone calcitonin and as an extracellular calcium sensor. Therefore, CTR dysfunction could lead to disorders of calcium metabolism associated with hypercalcemia, such as the Williams syndrome (WS). WS is a developmental disorder caused by a deletion at chromosome 7q11.23 that includes the elastin locus (ELN). We have mapped the CTR gene (CALCR) to chromosome band 7q21.3 by polymerase chain reaction and single-strand conformation analysis of somatic cell hybrids as well as fluorescence in situ hybridization (FISH) to metaphase chromosome spreads. Two-color FISH cohybridizing CTR and ELN probes confirmed that CALCR maps telomeric to ELN. Subsequent analysis of chromosome spreads from four WS patients revealed deletion of the ELN locus in all of them and normal hybridization of CTR probes to both chromosome 7 homologues, indicating that CALCR lies outside the deleted region.

    View details for Web of Science ID A1995RG53300071

    View details for PubMedID 7789182

  • Isolation of a novel gene mutated in Wiskott-Aldrich syndrome. Cell Derry, J. M., Ochs, H. D., FRANCKE, U. 1994; 79 (5): following 922-?

    View details for PubMedID 8001129

  • THE MOUSE PINK-EYED DILUTION GENE - ASSOCIATION WITH HYPOPIGMENTATION IN PRADER-WILLI AND ANGELMAN SYNDROMES AND WITH HUMAN OCA2 XVth International Pigment Cell Conference Brilliant, M. H., King, R., FRANCKE, U., Schuffenhauer, S., Meitinger, T., Gardner, J. M., DURHAMPIERRE, D., Nakatsu, Y. MUNKSGAARD INT PUBL LTD. 1994: 398–402

    Abstract

    Mutations at the mouse pink-eyed dilution locus, p, cause hypopigmentation. We have cloned the mouse p gene cDNA and the cDNA of its human counterpart, P. The region of mouse chromosome 7 containing the p locus is syntenic with human chromosome 15q11-q13, a region associated with Prader-Willi syndrome (PWS) and Angelman syndrome (AS), both of which involve profound imprinting effects. PWS patients lack sequences of paternal origin from 15q, whereas AS patients lack a maternal copy of an essential region from 15q. However, the critical regions for these syndromes are much smaller than the chromosomal region commonly deleted that often includes the P gene. Hypopigmentation in PWS and AS patients is correlated with deletions of one copy of the human P gene that is highly homologous with its mouse counterpart. A subset of PWS and AS patients also have OCA2. These patients lack one copy of the P gene in the context of a PWS or AS deletion, with a mutation in the remaining chromosomal homologue of the P gene. Mutations in both homologues of the P gene of OCA2 patients who do not have PWS or AS have also been detected.

    View details for Web of Science ID A1994QC40200005

    View details for PubMedID 7761348

  • IDENTIFICATION OF A NOVEL PATERNALLY EXPRESSED GENE IN THE PRADER-WILLI-SYNDROME REGION HUMAN MOLECULAR GENETICS Wevrick, R., Kerns, J. A., FRANCKE, U. 1994; 3 (10): 1877-1882

    Abstract

    We have isolated a novel gene from the Prader-Willi syndrome (PWS) smallest region of deletion overlap in proximal human chromosome 15q. IPW (Imprinted gene in the Prader-Willi syndrome region) was isolated using the direct selection method and yeast artificial chromosomes localized to the deletion region. IPW is spliced and polyadenylated but its longest open reading frame codes for only 45 amino acids, suggesting that it functions as an RNA, similar to H19 and XIST. The RNA is widely expressed in adult and fetal tissues and is found in the cytoplasmic fraction of human cells, which is also the case for the H19 non-translated RNA, but differs from the XIST RNA which is found predominantly in the nucleus. Using a sequence polymorphism, exclusive expression from the paternal allele in lymphoblasts and fibroblasts was demonstrated; monoallelic expression was found in fetal tissues. IPW is located about 150 kb distal to SNRPN, the only other known gene in the deletion interval, and about 50 kb proximal to the breakpoint of a translocation which defines the distal end of the PWS region and the proximal end of the Angelman syndrome (AS) region. As is the case with SNRPN, PWS patients with 15q11-q13 deletions do not express IPW, whereas expression is normal in Angelman syndrome patients. Lack of expression of IPW may contribute to the PWS phenotype directly. Alternatively, the mRNA product of IPW may play a role in the imprinting process, acting either on genes located proximally in the PWS region or distally in the AS region.

    View details for Web of Science ID A1994PN13800027

    View details for PubMedID 7849716

  • CDNA CLONING OF THE HUMAN MONOCARBOXYLATE TRANSPORTER-1 AND CHROMOSOMAL LOCALIZATION OF THE SLC16A1 LOCUS TO 1P13.2-P12 GENOMICS Garcia, C. K., Li, X., Luna, J., FRANCKE, U. 1994; 23 (2): 500-503

    Abstract

    Lactate and pyruvate are transported across cell membranes by monocarboxylate transporters (MCTs). Here, we use the recently cloned cDNA for hamster MCT1 to isolate cDNA and genomic clones for human MCT1. Comparison of the human and hamster amino acid sequences revealed that the proteins are 86% identical. The gene for human MCT1 (gene symbol, SLC16A1) was localized to human chromosome bands 1p13.2-p12 by PCR analysis of panels of human x rodent cell hybrid lines and by fluorescence chromosomal in situ hybridization.

    View details for Web of Science ID A1994PK06000027

    View details for PubMedID 7835905

  • QUANTITATIVE DIFFERENCES IN BIOSYNTHESIS AND EXTRACELLULAR DEPOSITION OF FIBRILLIN IN CULTURED FIBROBLASTS DISTINGUISH 5 GROUPS OF MARFAN-SYNDROME PATIENTS AND SUGGEST DISTINCT PATHOGENETIC MECHANISMS JOURNAL OF CLINICAL INVESTIGATION Aoyama, T., FRANCKE, U., Dietz, H. C., FURTHMAYR, H. 1994; 94 (1): 130-137

    Abstract

    Pulse-chase studies of [35S]cysteine-labeled fibrillin were performed on fibroblast strains from 55 patients with Marfan syndrome (MFS), including 13 with identified mutations in the fibrillin-1 gene and 10 controls. Quantitation of the soluble intracellular and insoluble extracellular fibrillin allowed discrimination of five groups. Groups I (n = 8) and II (n = 19) synthesize reduced amounts of normal-sized fibrillin, while synthesis is normal in groups III (n = 6), IV (n = 18), and V (n = 4). When extracellular fibrillin deposition is measured, groups I and III deposit between 35 and 70% of control values, groups II and IV < 35%, and group V > 70%. A deletion mutant with a low transcript level from the mutant allele and seven additional patients have the group I protein phenotype. Disease in these patients is caused by a reduction in microfibrils associated with either a null allele, an unstable transcript, or an altered fibrillin product synthesized in low amounts. In 68% of the MFS individuals (groups II and IV), a dominant negative effect is invoked as the main pathogenetic mechanism. Products made by the mutant allele in these fibroblasts are proposed to interfere with microfibril formation. Insertion, deletion, and exon skipping mutations, resulting in smaller fibrillin products, exhibit the group II phenotype. A truncated form of fibrillin of 60 kD was identified with specific fibrillin antibodies in one of the group II cell culture media. Seven of the nine known missense mutations, giving rise to abnormal, but normal-sized fibrillin molecules, are in group IV.

    View details for Web of Science ID A1994NW49600019

    View details for PubMedID 8040255

  • AFRICAN ORIGIN OF AN INTRAGENIC DELETION OF THE HUMAN-P GENE IN TYROSINASE POSITIVE OCULOCUTANEOUS ALBINISM NATURE GENETICS DURHAMPIERRE, D., Gardner, J. M., Nakatsu, Y., King, R. A., FRANCKE, U., Ching, A., Aquaron, R., DELMARMOL, V., Brilliant, M. H. 1994; 7 (2): 176-179

    Abstract

    Oculocutaneous albinism (OCA) is a genetically heterogeneous hypopigmentation disorder. One of the two major autosomal recessive forms involves the tyrosinase gene (OCA1), while the other form (OCA2) has recently been associated with alterations of the P gene on chromosome 15. OCA2 is about twice as common as OCA1 in African and African-American populations. We now describe an interstitial deletion that removes a single exon of the P gene. In a large family from an inbred population of tri-racial origin, all individuals with OCA2 were found to be homozygous for this allele. Moreover, the same mutant P allele was detected in several unrelated African American individuals with OCA2, but not in Caucasians with OCA2. The detection of the same allele in two unrelated Africans with OCA2 indicates an African origin for this allele.

    View details for Web of Science ID A1994NQ03700018

    View details for PubMedID 7920637

  • MARFANS-SYNDROME AND OTHER DISORDERS OF FIBRILLIN NEW ENGLAND JOURNAL OF MEDICINE FRANCKE, U., FURTHMAYR, H. 1994; 330 (19): 1384-1385

    View details for Web of Science ID A1994NK75300011

    View details for PubMedID 8152453

  • Receptor mutations and haplotypes in growth hormone receptor deficiency: a global survey and identification of the Ecuadorean E180splice mutation in an oriental Jewish patient. Acta paediatrica Berg, M. A., Peoples, R., Pérez-Jurado, L., Guevara-Aguirre, J., Rosenbloom, A. L., Laron, Z., MILNER, R. D., FRANCKE, U. 1994; 399: 112-114

    Abstract

    Eight different mutations were detected in the growth hormone (GH) receptor gene of patients with inherited GH receptor deficiency (GHRD; Laron syndrome) from five continents. All the mutations are located in the extracellular domain of the receptor and are predicted to cause gross structural abnormalities and non-functional receptor molecules. They include three nucleotide changes in the coding region causing translational stop signals, including the newly identified E183X mutation; two nucleotide changes in introns that affect splice junctions; two dinucleotide deletions that result in stop codons downstream; and one single nucleotide change that activates a donor splice site within an exon and results in a transcript missing 24 nucleotides. This latter mutation (E180splice) was first identified in a cohort of patients with GHRD from southern Ecuador. Based on the fact that the E180splice mutation generates a new cleavage site for the restriction enzyme MnlI, a simple diagnostic test has been developed that can be carried out on dried blood spots collected on filter paper. A total of 55 affected individuals from Ecuador has been found to be homozygous for this mutation. Asymptomatic carriers can also be detected, and 104 of 150 individuals screened were found to be carriers. Using this test, the E180splice mutation has recently been detected in one of two oriental Jewish patients from Israel.

    View details for PubMedID 7949594

  • CLONING OF MITF, THE HUMAN HOMOLOG OF THE MOUSE MICROPHTHALMIA GENE AND ASSIGNMENT TO CHROMOSOME 3P14.1 - P12.3 HUMAN MOLECULAR GENETICS Tachibana, M., PEREZJURADO, L. A., Nakayama, A., Hodgkinson, C. A., Li, X., Schneider, M., Miki, T., FEX, J., FRANCKE, U., Arnheiter, H. 1994; 3 (4): 553-557

    Abstract

    The mouse microphthalmia (mi) gene encodes a basic-helix-loop-helix-zipper protein whose mutations may lead to loss of pigmentation in the eye, inner ear and skin, and to reduced eye size and early onset deafness. Mice with mutations at mi serve as models for human pigment disturbances in skin and eye that may be combined with sensorineural deafness. We have now obtained cDNA and genomic clones of the human homolog of mouse mi, identified a restriction fragment length polymorphism in the gene, and mapped the gene by somatic cell hybrid and fluorescence in situ hybridization techniques to a region of human chromosome 3 that shows a disrupted syntenic conservation with the region on mouse chromosome 6 to which mi maps. These studies will help to verify if any of the hereditary pigment disturbances in humans are due to mutations in this gene.

    View details for Web of Science ID A1994NH45600004

    View details for PubMedID 8069297

  • GENE FOR A TISSUE-SPECIFIC TRANSCRIPTIONAL ACTIVATOR (EBF OR OLF-1), EXPRESSED IN EARLY B-LYMPHOCYTES, ADIPOCYTES, AND OLFACTORY NEURONS, IS LOCATED ON HUMAN CHROMOSOME-5, BAND-Q34, AND PROXIMAL MOUSE CHROMOSOME-11 MAMMALIAN GENOME MILATOVICH, A., Qiu, R. G., Grosschedl, R., FRANCKE, U. 1994; 5 (4): 211-215

    Abstract

    Murine B lymphocytes, adipocytes, and olfactory neurons contain a DNA-binding protein that participates in the regulation of genes encoding tissue-specific components of signal transduction. Purification and cloning of this protein, termed early B-cell factor (EBF), from murine B lymphocytes and independent cloning of a protein, termed Olf-1, from olfactory neuronal cells revealed virtual complete amino acid sequence identity between these proteins. As a first step towards identifying a human genetic disorder or mouse mutation for which EBF could be a candidate gene, we have chromosomally mapped the corresponding locus in both species. By Southern hybridization analyses of somatic cell hybrid panels with murine cDNA probe, fluorescence chromosomal in situ hybridization (FISH) of human genomic clones, and analysis of recombinant inbred mouse strains, we have found single sites for EBF homologous sequences on human Chromosome (Chr) 5, band q34, and on proximal mouse Chr 11, in an evolutionarily conserved region.

    View details for Web of Science ID A1994ND31300004

    View details for PubMedID 8012110

  • EXCLUSION OF GROWTH-HORMONE (GH)-RELEASING HORMONE GENE-MUTATIONS IN FAMILIAL ISOLATED GH DEFICIENCY BY LINKAGE AND SINGLE-STRAND CONFORMATION ANALYSIS JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM Jurado, L. A., Phillips, J. A., FRANCKE, U. 1994; 78 (3): 622-628

    Abstract

    The molecular basis and the locus responsible for most familial cases of isolated GH deficiency (IGHD) are still unknown. The GH-releasing hormone (GHRH) gene has been evaluated as a possible candidate in 23 unrelated families with IGHD, 14 of whom were classified as having autosomal recessive IGHD type IB and 9 of whom had autosomal dominant IGHD type II. Three highly polymorphic microsatellites (dinucleotide repeats), mapped close to GHRH on chromosome 20 by previous linkage studies, were analyzed as markers for the GHRH locus. All available family members were genotyped for D20S44 [no recombination with GHRH at a LOD (logarithm of the odds) score of 3.6]. Noninformative families were also genotyped for D20S45 and/or D20S54 (located at approximately 1 and 3 centiMorgan of genetic distance from GHRH, respectively). Twenty families were informative for linkage analysis with 1 or more of these markers. We found at least 1 obligate recombinant with discordance between phenotype and genotype in 19 of the 23 families (83%). There is only a very small chance (1-3% or less) that the discordances observed are due to recombination between the GHRH locus and the marker tested. Concordant segregation was seen in only 1 type IB family (4%). When probands from this and the 3 noninformative families were screened for sequence variants in the 5 exons of the GHRH gene by single strand conformation analysis, no abnormal patterns were observed. We conclude that mutations responsible for IGHD are not within or near the structural gene for GHRH on chromosome 20 in the 23 families studied. As linkage to the GH-1 gene has also been previously excluded in 65% of these families, mutations in a locus or loci unlinked to GH-1 and GHRH must be responsible for the majority of these IGHD families.

    View details for Web of Science ID A1994MZ30700020

    View details for PubMedID 8126133

  • CHROMOSOME LOCALIZATIONS OF GENES FOR 5 CAMP-SPECIFIC PHOSPHODIESTERASES IN MAN AND MOUSE SOMATIC CELL AND MOLECULAR GENETICS MILATOVICH, A., Bolger, G., Michaeli, T., FRANCKE, U. 1994; 20 (2): 75-86

    Abstract

    Cyclic nucleotides are important second messengers that mediate a number of cellular responses to external signals. Cyclic nucleotide phosphodiesterases play a role in signal transduction by regulating the cellular concentrations of these messengers. Here, we have applied Southern analyses of somatic cell hybrid lines and of recombinant inbred (RI) mouse strains as well as fluorescence chromosomal in situ hybridization (FISH) to chromosomally localize five cAMP-specific nucleotide phosphodiesterase genes in human and mouse. Genes DPDE1, DPDE2, DPDE3, and DPDE4 that share sequence homology with the Drosophila dunce gene were assigned to human chromosome 19 (DPDE1 and DPDE2), 5q12 (DPDE3), and 1p31 (DPDE4) and to mouse chromosomes 8, 9, 13, and 4, respectively. The high-affinity cAMP-specific phosphodiesterase gene (HCP1) was mapped to human chromosome 8q13-q22. Since these genes are potential candidates for involvement in psychiatric or behavioral disorders, knowledge of their chromosomal localizations will facilitate the discovery of their association with disease genes as they are being mapped by linkage studies.

    View details for Web of Science ID A1994NU89100001

    View details for PubMedID 8009369

  • SINGLE NUCLEOTIDE DIMORPHISM IN THE TRANSCRIBED REGION OF THE SNRPN GENE AT 15Q12 HUMAN MOLECULAR GENETICS Giacalone, J., FRANCKE, U. 1994; 3 (2): 379-379

    View details for Web of Science ID A1994MY58300033

    View details for PubMedID 7911699

  • STRUCTURE AND LOCALIZATION ON THE X-CHROMOSOME OF THE GENE CODING FOR THE HUMAN FILOPODIAL PROTEIN MOESIN (MSN) GENOMICS Wilgenbus, K. K., Hsieh, C. L., Lankes, W. T., MILATOVICH, A., FRANCKE, U., FURTHMAYR, H. 1994; 19 (2): 326-333

    Abstract

    Moesin is a member of a recently discovered family of closely related proteins that includes ezrin, radixin, and merlin. It is widely expressed in different tissues and cells and has been localized to filopodia and other membranous protrusions that are important for cell-cell recognition and signaling and cell movement. Here, we have localized the coding gene (MSN) to Xq11.2-q12 by Southern and Western blot analyses of Chinese hamster x human somatic cell hybrids and by fluorescence chromosomal in situ hybridization. Moesin-like sequences were identified on chromosomes 5 and 6. The murine Msn locus was mapped to the X chromosome as well by studying a rodent x mouse hybrid panel. The structure of the human moesin gene has been determined. The 12 exons are distributed over > 30 kb, and the exon/intron junctions demarcate individual highly conserved domains. Primer extension analysis revealed two major start transcription sites, 184 and 133 bp upstream of the initiation codon. The 5'-flanking region is GC-rich, lacks a TATA box, and contains four SP1 and one AP1 binding sites.

    View details for Web of Science ID A1994NA60300017

    View details for PubMedID 8188263

  • LOCALIZATION OF THE CAMKG GENE ENCODING GAMMA-ISOFORMS OF MULTIFUNCTIONAL CALCIUM/CALMODULIN-DEPENDENT PROTEIN-KINASE (CAM KINASE) TO HUMAN-CHROMOSOME-10 BAND Q22 AND MOUSE CHROMOSOME-14 CYTOGENETICS AND CELL GENETICS Li, X., Nghiem, P., Schulman, H., FRANCKE, U. 1994; 66 (2): 113-116

    Abstract

    Multifunctional calcium/calmodulin-dependent protein kinase (CaM kinase) is an enzyme mediating calcium-dependent signal transduction pathways. CaM kinase exists in a variety of isoforms, each with a distinct tissue-specific expression pattern, that enables the kinase to regulate multiple functions in mammalian systems. Here we report the chromosomal localization of the previously cloned human gamma-CaM kinase gene (CAMKG). By using a mapping panel of human x Chinese hamster somatic cell hybrid lines and fluorescence in situ hybridization, we have assigned human CAMKG to chromosome 10q22. We have partially cloned the murine gamma-CaM kinase gene and mapped it Camkg to mouse chromosome 14 by analyzing a panel of mouse x rodent somatic cell hybrid lines. A recessive gene, asa, implicated in the control of autoimmune response, is located within the predicted region for Camkg.

    View details for Web of Science ID A1994MV18600009

    View details for PubMedID 8287681

  • AUTOSOMAL RECESSIVE NEUROMUSCULAR DISORDER IN A TRANSGENIC LINE OF MICE JOURNAL OF NEUROSCIENCE Kelly, D., Chancellor, K., MILATOVICH, A., FRANCKE, U., Suzuki, K., Popko, B. 1994; 14 (1): 198-207

    Abstract

    We have generated a line of transgenic mice that when homozygous for the transgene develop a severe, adult-onset neuromuscular disorder. This mutation is likely the result of the insertional inactivation of an endogenous gene by the transgene integration. The mutant mice have a gait abnormality with stiffened and/or splayed hind legs, and adopt a hunched posture with some exhibiting kyphosis of the thoracic spine. These symptoms progress gradually to severe motor dysfunction. Pathologic changes were found in skeletal muscle and peripheral nerve of the mutant animals. In young mice the muscles from both upper and lower extremities show necrosis and phagocytosis. In older mice, regeneration with muscle fiber splitting, internally located nuclei, and variable fiber size are conspicuous features. Interactions between Schwann cells and axons also appear disrupted in these animals. Although many peripheral axons are well myelinated, the nerve and nerve roots contain very large bundles of juxtaposed, bare axons, reminiscent of Schwann cell-axon interactions in early development. Within these bundles there are axons large enough to be myelinated. The relationship between the pathologic changes in the muscles and nerves is not clear. The phenotypic abnormalities of these animals resemble those that occur in the spontaneous mouse mutants dystrophia muscularis and myodystrophy. Nevertheless, the chromosomal position of the transgene integration site, which was mapped by fluorescent in situ hybridization to chromosome 11, indicates that this disorder represents a new neuromuscular mutation.

    View details for Web of Science ID A1994MQ11800017

    View details for PubMedID 7506762

  • REGIONAL LOCALIZATION OF 56 NEW HUMAN CHROMOSOME-18 SPECIFIC YEAST ARTIFICIAL CHROMOSOMES CYTOGENETICS AND CELL GENETICS Chang, E., Luna, J., Giacalone, J., Uyar, D., Silverman, G. A., FRANCKE, U. 1994; 65 (1-2): 136-139

    Abstract

    Fifty-six chromosome 18-specific yeast artificial chromosomes (YAC) were isolated from a human monochromosomal somatic cell hybrid DNA library. Fluorescence in situ hybridization revealed that the clones were evenly distributed throughout the chromosome. The clones, with an average insert size of 250 kb, cover about 18% of the euchromatic part of chromosome 18. Of 90 STS markers tested, 17 were represented in this YAC collection. Together with our previously reported set of unique chromosome 18-specific YACs, we now have available 111 regionally mapped, essentially nonchimeric, clones that provide more than 30% coverage and form a framework for the complete physical map.

    View details for Web of Science ID A1994ME40400087

    View details for PubMedID 8404067

  • A RADIATION HYBRID MAP OF HUMAN CHROMOSOME-18 CYTOGENETICS AND CELL GENETICS FRANCKE, U., Chang, E., COMEAU, K., Geigl, E. M., Giacalone, J., Li, X., Luna, J., Moon, A., Welch, S., Wilgenbus, P. 1994; 66 (3): 196-213

    Abstract

    A human x Chinese hamster (CH) somatic cell hybrid subclone deficient in HPRT and containing only human chromosome 18 was irradiated with 7000 rad and fused to a thymidine kinase deficient CH cell line. Radiation-rescued hybrid cell lines, selected in HAT medium, were analyzed for human DNA with human interspersed-repeat sequence primers. Size and number of human chromosome fragments retained in a subset of hybrids were determined by FISH. A panel of 98 radiation hybrids (RH) was selected and analyzed for 90 chromosome 18-specific STSs by PCR, and for the D18Z1 centromeric marker by Southern blotting. STSs were developed from previously mapped RFLP loci and from published sequences. In addition, 32 novel STSs were generated from an 18-specific lambda library and from 18-specific YACs previously localized to chromosome bands by FISH. Marker retention frequency varied from 8-65% with an average of 24%. In selected RH the STS typing data were correlated with the chromosome 18 regions retained using 'reverse FISH' of IRS-PCR products from the RH to normal metaphase chromosomes. The order and intermarker distances of loci were determined using two-point and multipoint maximum likelihood methods. The resulting RH map covers most of chromosome 18 with four groups of tightly linked markers and three regions of loosely linked markers, one around the centromere and two on the long arm. More than a third of the markers are polymorphic and allow integration with the linkage map. This RH map provides a framework for establishing a clone contig of the entire chromosome 18.

    View details for Web of Science ID A1994NC70200011

    View details for PubMedID 8125019

  • DIGITIZED AND DIFFERENTIALLY SHADED HUMAN-CHROMOSOME IDEOGRAMS FOR GENOMIC APPLICATIONS CYTOGENETICS AND CELL GENETICS FRANCKE, U. 1994; 65 (3): 206-218

    Abstract

    Human chromosome ideograms that depict 850 bands, numbered in agreement with ISCN (1981) nomenclature but based on actual measurements of band sizes and differentiated by five shades of staining intensity, have been digitized using commercially available graphics software. They provide for more accurate estimates of distances within chromosome arms, DNA content of chromosome bands and correlation between cytogenetic, molecular and genetic linkage maps than the standard ISCN ideograms.

    View details for Web of Science ID A1994MJ49100035

    View details for PubMedID 8222762

  • STRUCTURE, CHROMOSOMAL LOCALIZATION, AND EXPRESSION PATTERN OF THE MURINE MAGP GENE JOURNAL OF BIOLOGICAL CHEMISTRY Chen, Y. P., Faraco, J., Yin, W. S., GERMILLER, J., FRANCKE, U., Bonadio, J. 1993; 268 (36): 27381-27389

    Abstract

    The microfibril-associated glycoprotein (MAGP) was recently established as a discrete constituent of 10-nm microfibrils. We have characterized the primary structure of the mouse transcript, the structure and chromosomal localization of the murine gene, and the developmental pattern of gene expression. The transcript consists of 1,037 base pairs as determined by cDNA cloning, Northern blot analysis, S1 nuclease mapping, and primer extension mapping. Using a cDNA fragment as a probe, we isolated a single genomic clone that contained the entire mouse gene. Analysis of this clone indicated that Magp is fragmented into 9 exons, with the initiator Met codon located in exon 2. As determined by analysis of somatic cell hybrid lines and by fluorescence in situ hybridization, the mouse gene was mapped to chromosome 4 at a location corresponding to region D3-E1. Genomic sequence immediately upstream of the transcription start site was found to be GC-rich but lacked TATA or CCAAT boxes as well as other cis-acting motifs known to regulate transcription. Promoters of this type are usually found in genes that exhibit broad temporal and spatial patterns of expression. Consistent with this idea, the Magp transcript appeared to be the widespread product of mesenchymal/connective tissue cells throughout mouse development. This study presents the first comprehensive evaluation of microfibril gene expression during mammalian development.

    View details for Web of Science ID A1993MM26200078

    View details for PubMedID 8262979

  • FIBRILLIN GENES MAP TO REGIONS OF CONSERVED MOUSE HUMAN SYNTENY ON MOUSE CHROMOSOME-2 AND CHROMOSOME-18 GENOMICS Li, X., Pereira, L., Zhang, H., Sanguineti, C., Ramirez, F., Bonadio, J., FRANCKE, U. 1993; 18 (3): 667-672

    Abstract

    Fibrillin proteins are major structural components of the 10-nm microfibrils found in elastic and nonelastic connective tissues. Previous studies have mapped the human genes for two fibrillins to chromosome bands 15q21 (FBN1) and 5q23-q31 (FBN2) and have demonstrated that FBN1 mutations are associated with Marfan syndrome, while FBN2 is linked to the gene for congenital contractural arachnodactyly. Here, we report the isolation of genomic clones of the corresponding mouse fibrillin genes (Fbn-1 and Fbn-2). By analyzing a mapping panel of mouse x rodent somatic hybrid cell lines, we have assigned the Fbn-1 gene to mouse chromosome 2 and the Fbn-2 gene to mouse chromosome 18. We then sublocalized the fibrillin genes to bands 2F (Fbn-1) and 18D-E1 (Fbn-2) by fluorescence in situ hybridization. These regions are known to exhibit conserved synteny with the regions on human chromosomes 15 and 5 that carry the homologous human fibrillin genes. In addition, the Fbn-1 gene maps in the vicinity of the gene for a connective tissue disorder on mouse chromosome 2 called Tight-skin (Tsk).

    View details for Web of Science ID A1993MP48000028

    View details for PubMedID 8307578

  • MISSENSE MUTATIONS IMPAIR INTRACELLULAR PROCESSING OF FIBRILLIN AND MICROFIBRIL ASSEMBLY IN MARFAN-SYNDROME HUMAN MOLECULAR GENETICS Aoyama, T., Tynan, K., Dietz, H. C., FRANCKE, U., FURTHMAYR, H. 1993; 2 (12): 2135-2140

    Abstract

    Dermal fibroblasts from nine Marfan syndrome patients with missense mutations in the fibrillin-1 gene (FBN1) produced nearly normal amounts of fibrillin as determined by quantitative pulse-chase experiments. However, six of the seven mutations involving substitutions of highly conserved cysteine residues exhibited lower rates of intracellular transport and secretion. This effect is likely due to improper folding, since intracellular fibrillin processing was also affected by the reducing agent dithiothreitol. Normal secretion patterns were seen in three mutations that either change the conformation of EGF-like domains or change consensus amino acids required for Ca(++)-binding. In all nine fibroblasts strains, however, the deposition of fibrillin in the extracellular matrix was reduced to 50% of normal in two and to less than 30% in seven of the nine samples studied. The protein alterations caused by these missense mutations are associated with moderate to severe features of Marfan syndrome and a dominant negative mechanism is suggested to play a major role in their pathogenesis.

    View details for Web of Science ID A1993MP21300023

    View details for PubMedID 8111384

  • ROBERTS-SYNDROME - A REVIEW OF 100 CASES AND A NEW RATING SYSTEM FOR SEVERITY AMERICAN JOURNAL OF MEDICAL GENETICS VANDENBERG, D. J., FRANCKE, U. 1993; 47 (7): 1104-1123

    Abstract

    Roberts syndrome (RS) is a rare genetic disorder characterized by pre- and postnatal growth retardation, limb defects, and craniofacial anomalies. Affected persons have varying degrees of malformations involving symmetric reduction in the number of digits, and length or presence of bones in the arms and legs. Craniofacial malformations involve hypertelorism, hypoplastic nasal alae, and a high incidence of cleft lip and palate. Familial and sporadic cases have been reported consistent with an autosomal recessive mode of inheritance. Mitotic cells from many individuals with RS display a characteristic cytogenetic phenomenon consisting of repulsion of heterochromatic regions near centromeres, particularly of chromosomes 1, 9, 16, and splaying of the short arms of the acrocentrics and of the distal Yq. Mitosis in RS cells is abnormal in metaphase duration and anaphase progression. Specifically, anaphase figures show a higher degree of chromosomes that are outlying, lagging, or prematurely advancing toward the poles compared to normal controls. RS cells have abnormal nuclear morphology and also show a higher frequency of micronucleation than normal cells, presumably as a result of the abnormal mitotic events during anaphase. Therefore, RS has been interpreted as a human mitotic mutation syndrome which leads to secondary developmental defects. This report reviews 100 cases of RS, summarizes the phenotypic, genetic, cytogenetic, and cell biology findings in Roberts syndrome, and introduces the RS Rating for quantitating severity.

    View details for Web of Science ID A1993ME48700034

    View details for PubMedID 8291532

  • MUTATION SCREENING OF COMPLETE FIBRILLIN-1 CODING SEQUENCE - REPORT OF 5 NEW MUTATIONS, INCLUDING 2 IN 8-CYSTEINE DOMAINS HUMAN MOLECULAR GENETICS Tynan, K., COMEAU, K., Pearson, M., Wilgenbus, P., Levitt, D., Gasner, C., Berg, M. A., Miller, D. C., FRANCKE, U. 1993; 2 (11): 1813-1821

    Abstract

    Marfan syndrome (MFS) is an autosomal dominantly inherited connective tissue disorder characterized by cardiovascular, ocular and skeletal manifestations. Previously, mutations in the fibrillin-1 gene on chromosome 15 (FBN1) have been reported to cause MFS. We have now screened 44 probands with MFS or related phenotypes for alterations in the entire fibrillin coding sequence (9.3 kb) by single strand conformation analysis. We report four unique mutations in the fibrillin gene of unrelated MFS patients. One is a 17 bp deletion and three are missense mutations, two of which involve 8-cysteine motifs. Another missense mutation was found in two unrelated individuals with annuloaortic ectasia but was also present in unaffected relatives and controls from various ethnic backgrounds. By using allele-specific oligonucleotide hybridization, we screened 65 unrelated MFS patients, 29 patients with related phenotypes and 84 control individuals for these mutations as well as for a previously reported mutation and two polymorphisms. Our results suggest that most MFS families carry unique mutations and that the fibrillin genotype is not the sole determinant of the connective tissue phenotype.

    View details for PubMedID 8281141

  • GENOMIC DISTRIBUTION AND TRANSCRIPTION OF SOLITARY HERV-K LTRS GENOMICS LEIBMOSCH, C., Haltmeier, M., Werner, T., Geigl, E. M., BRACKWERNER, R., FRANCKE, U., Erfle, V., Hehlmann, R. 1993; 18 (2): 261-269

    Abstract

    The human genome contains a family of endogenous retroviruses, HERV-K, with sequence homology to the B-type mouse mammary tumor virus. We have now identified a single HERV-K LTR within the C-type-related human retroviral element S71. The HERV-K LTR is located in the antisense direction between the S71 gag and the pol gene, replacing the 5' half of S71 pol. A number of HERV-K LTR-related cDNA clones were detected by screening various human cDNA libraries with an S71 HERV-K LTR probe, indicating abundant transcription of HERV-K-related LTRs in human tissues. Sequence analysis of four cDNA clones revealed LTR sequences with a nucleotide identity of 70 to 90% with HERV-K10 LTR. Some HERV-K-related LTR sequences contain potential short open reading frames. The analyzed cDNA clones do not harbor any retroviral sequences other than those related to HERV-K LTRs. However, most of the solitary LTRs were found to be coexpressed with cellular sequences. Transcription of these LTRs is probably directed by external cellular promoters. We show that HERV-KLTR-like sequences entered the primate genome about 33-40 million years ago. We estimate the human genome to contain about 25,000 copies of HERV-K-related LTRs, which are distributed over most human chromosomes in an irregular manner.

    View details for Web of Science ID A1993MG99300013

    View details for PubMedID 8288228

  • GENE FOR THE ALPHA-SUBUNIT OF THE HUMAN INTERLEUKIN-3 RECEPTOR (IL3RA) LOCALIZED TO THE X-Y PSEUDOAUTOSOMAL REGION AMERICAN JOURNAL OF HUMAN GENETICS MILATOVICH, A., Kitamura, T., Miyajima, A., FRANCKE, U. 1993; 53 (5): 1146-1153

    Abstract

    Interleukin-3 (IL3) and granulocyte/macrophage colony-stimulating factor (CSF2) stimulate proliferation and differentiation of various hemopoietic cell types. As is characteristic of the cytokine receptor family, the receptors for these proteins comprise alpha- and beta-subunits. While IL3 and CSF2 receptors each have unique alpha- subunits, they share a common beta-subunit. By Southern analysis of somatic cell hybrid panels, pulsed-field gel electrophoresis (PFGE), and fluorescence chromosomal in situ hybridization, we have mapped the cloned sequence for the IL3 receptor alpha (IL3RA) to the X-Y pseudoautosomal region at bands Xp22.3 and Yp11.3, near the gene for the alpha-subunit of the CSF2 receptor (CSF2RA). The CSF2RA and IL3RA genes are so close that their order could not be determined by two-color interphase in situ hybridization. They share PFGE fragments generated by different restriction enzymes down to the 50-100-kb size range. Pseudoautosomal inheritance was demonstrated by an EcoRI RFLP detected with the IL3RA cDNA probe.

    View details for Web of Science ID A1993MC33300019

    View details for PubMedID 8213838

  • THE HUMAN HOMOLOG OF THE GLOMERULOSCLEROSIS GENE MPV17 - STRUCTURE AND GENOMIC ORGANIZATION HUMAN MOLECULAR GENETICS Karasawa, M., Zwacka, R. M., Reuter, A., Fink, T., Hsieh, C. L., Lichter, P., FRANCKE, U., Weiher, H. 1993; 2 (11): 1829-1834

    Abstract

    Mice carrying a retroviral insert in both alleles of the Mpv17 gene develop glomerulosclerosis and nephrotic syndrome at young age. Thus, the Mpv17 gene is a recessive disease gene in mice and this mouse strain is a potential animal model for glomerular diseases in man. We here describe the isolation and analysis of a human homolog of this gene. By interspecies hybridisation cDNA clones representing a single RNA species were isolated from human liver. Sequence analysis revealed over 90% identify in a region coding for a protein of 176 amino acids and unknown function in both species. Cloning of the genomic locus revealed a single copy gene which we mapped to the short arm of chromosome 2 at band 2p23-p21. Determination of the intron-exon structure and the junction sequences enabled us to establish a PCR based procedure to isolate the coding region from human genomic DNA. Thus, it is now possible to analyse patients suffering from candidate diseases on the basis of a blood sample if biopsy material is not available.

    View details for Web of Science ID A1993MG82700012

    View details for PubMedID 8281143

  • HUMAN DYSTROGLYCAN - SKELETAL-MUSCLE CDNA, GENOMIC STRUCTURE, ORIGIN OF TISSUE-SPECIFIC ISOFORMS AND CHROMOSOMAL LOCALIZATION HUMAN MOLECULAR GENETICS IBRAGHIMOVBESKROVNAYA, O., MILATOVICH, A., Ozcelik, T., Yang, B., KOEPNICK, K., FRANCKE, U., Campbell, K. P. 1993; 2 (10): 1651-1657

    Abstract

    Dystroglycan is a novel laminin binding component of the dystrophin-glycoprotein complex which provides a linkage between the subsarcolemmal cytoskeleton and the extracellular matrix. Here we report the cDNA and genomic structure of human dystroglycan. The human dystroglycan is encoded by a single gene (DAG1) mapped to chromosome 3 band p21. The coding sequence is organized into two exons, separated by a large intron. The predicted amino acid sequence of human and rabbit dystroglycan are 93% identical with predicted glycosylation sites being conserved. Human dystroglycan is expressed in a variety of fetal and adult tissues. Our data suggest that muscle and non-muscle isoforms of dystroglycan differ by carbohydrate moieties but not protein sequence. Therefore, we hypothesize that variable glycosylation of the conserved protein core might modulate laminin binding. The relationship of dystroglycan to human diseases is discussed.

    View details for Web of Science ID A1993MC22300021

    View details for PubMedID 8268918

  • CDNA CLONING OF THE 2 SUBUNITS OF HUMAN CAAX FARNESYLTRANSFERASE AND CHROMOSOMAL MAPPING OF FNTA AND FNTB LOCI AND RELATED SEQUENCES GENOMICS Andres, D. A., MILATOVICH, A., Ozcelik, T., Wenzlau, J. M., BROWN, M. S., Goldstein, J. L., FRANCKE, U. 1993; 18 (1): 105-112

    Abstract

    The CAAX farnesyltransferase is a heterodimeric enzyme that attaches a farnesyl group to a single cysteine in several cellular proteins. Substrates include the p21ras proteins, nuclear lamins, and several retinal proteins, all of which end with a "CAAXbox," where C is cysteine, A is an aliphatic amino acid, and X is methionine or serine. Full-length cDNAs for the alpha and beta subunits of the rat farnesyltransferase have been cloned, and both have been shown to be essential for catalytic activity. Here we have used the rat cDNAs to clone cDNAs for the human alpha and beta subunits. Comparison of the human and rat amino acid sequences revealed a remarkable degree of conservation (93% identity for the alpha subunit and 96% identity for the beta subunit). The functional genes for the alpha and beta subunits of human farnesyltransferase (gene symbols, FNTA and FNTB) were localized to human chromosome bands 8p22-q11 and 14q23-q24, respectively, by Southern blot hybridization and PCR analyses of panels of human x Chinese hamster somatic cell hybrid lines and by fluorescence chromosomal in situ hybridization. We also found several related farnesyltransferase genes. FNTAL1 was assigned to 11q13.4-q14.1, FNTAL2 to chromosome 13, and FNTBL1 to chromosome 9.

    View details for Web of Science ID A1993MB01900014

    View details for PubMedID 8276393

  • GENETIC-HETEROGENEITY IN LARON SYNDROME 15TH INTERNATIONAL SYMP ON GROWTH AND GROWTH DISORDER FRANCKE, U., Berg, M. A. SCANDINAVIAN UNIVERSITY PRESS. 1993: 3–7

    View details for Web of Science ID A1993MB05300002

    View details for PubMedID 8219474

  • GENETIC-HETEROGENEITY IN LARON SYNDROME - DISCUSSION ACTA PAEDIATRICA Wilton, P., FRANCKE, U., POSTELVINAY, M. C., BARTSOUCAS, C., Hughes, I. A. 1993; 82: 8-8
  • A SATELLITED 4Q IDENTIFIED IN AMNIOTIC-FLUID CELLS Miller, I., SONGSTER, S., Fontana, S., FRANCKE, U., Hsieh, C. L. CELL PRESS. 1993: 1586–86
  • AORTIC-ANEURYSM IN A FAMILY WITH A GLYCINE-TO-SERINE SUBSTITUTION IN THE FBN-1 GENE Berg, M. A., Tynan, K., Aoyama, T., Gasner, C., COMEAU, K., Pearson, M., Levitt, D., Alcorn, D., Miller, C., FURTHMAYR, H., FRANCKE, U. CELL PRESS. 1993: 151–51
  • The Second International Symposium on the Marfan Syndrome. American journal of medical genetics Pyeritz, R. E., FRANCKE, U. 1993; 47 (1): 127-135

    View details for PubMedID 8103631

  • GENERATION OF A HUMAN CHROMOSOME-18-SPECIFIC YAC CLONE COLLECTION AND MAPPING OF 55 UNIQUE YACS BY FISH AND FINGERPRINTING GENOMICS Chang, E., Welch, S., Luna, J., Giacalone, J., FRANCKE, U. 1993; 17 (2): 393-402

    Abstract

    A yeast artificial chromosome (YAC) library was constructed from a somatic cell hybrid line in which the human chromosome content had been reduced by repeated subcloning to one or two copies of chromosome 18. Screening of 4700 primary yeast transformants generated 74 clones containing a YAC with a human DNA insert averaging 190 kb in size. The human YACs were localized to subregions of chromosome 18 by in situ hybridization of biotin-labeled Alu-PCR products obtained using total yeast DNA as a template. Comparisons of interspersed repetitive sequence-PCR and restriction fragment fingerprint patterns identified five sets of identical and three sets of overlapping YACs. Dual-label fluorescence in situ hybridization interphase mapping was used to determine the order of some nonoverlapping YACs. STS (sequence-tagged site) content mapping was carried out with PCR primers for 56 chromosome 18-specific markers. The identification of YACs containing four known genes--encoding the pituitary adenylate cyclase activating polypeptide (PA-CAP), the myelin basic protein (MBP), ferrochelatase (FECH), and SSAV1, an endogenous retroviral element related to the SSAV virus--provides a precise cytological map position for the respective loci. Our final collection of 55 randomly isolated, unique, and regionally localized YACs (D18S107-D18S161) is distributed over the entire chromosome and collectively covers approximately 12 Mb, i.e., 16% of the estimated 77 Mb of DNA in euchromatin of chromosome 18. These YACs provide reagents for the isolation of genes in these regions and represent nucleation points for the generation of STS to increase coverage of the chromosome by YAC contigs.

    View details for Web of Science ID A1993LP81500018

    View details for PubMedID 7691716

  • SENSITIVITY OF ROBERTS-SYNDROME CELLS TO GAMMA-RADIATION, MITOMYCIN-C, AND PROTEIN-SYNTHESIS INHIBITORS SOMATIC CELL AND MOLECULAR GENETICS VANDENBERG, D. J., FRANCKE, U. 1993; 19 (4): 377-392

    Abstract

    Roberts syndrome (RS) is a rare autosomal recessive disorder characterized by pre- and postnatal growth retardation, limb reduction abnormalities, and craniofacial anomalies. Mitotic chromosomes from RS individuals display repulsion of heterochromatin regions or centromere splitting, leading to a railroad-track appearance of mitotic chromosomes. Abnormalities in metaphase duration, anaphase progression, nuclear morphology, and increased frequency of micronucleation have been reported in RS cells. Cells from RS heterozygotes are normal in these respects, and in vitro complementation of the defects in somatic cell hybrids has been reported. Therefore, in preparation for the isolation of cDNAs that complement the RS defect, we investigated various drug treatments to identify an agent that specifically involves the growth of RS cells. Based on the cytogenetic and cell biologic findings, we chose agents that increase micronucleation or inhibit protein synthesis. We found that RS cells are hypersensitive to gamma radiation, mitomycin C, G418 and hygromycin B, but not to colcemid or streptonigrin when compared to normal cells. DNA content and cell viability analysis confirmed that the sensitivity to gamma irradiation was primarily due to increased cell death.

    View details for Web of Science ID A1993LZ82100007

    View details for PubMedID 8211379

  • STRUCTURAL COMPARISON AND CHROMOSOMAL LOCALIZATION OF THE HUMAN AND MOUSE IL-13 GENES JOURNAL OF IMMUNOLOGY McKenzie, A. N., Li, X., Largaespada, D. A., Sato, A., Kaneda, A., Zurawski, S. M., Doyle, E. L., MILATOVICH, A., FRANCKE, U., Copeland, N. G., Jenkins, N. A., Zurawski, G. 1993; 150 (12): 5436-5444

    Abstract

    The genomic structure of the recently described cytokine IL-13 has been determined for both human and mouse genes. The nucleotide sequence of a 4.6-kb DNA segment of the human gene is described. The human IL-13 gene (IL13) occurs as a single copy in the haploid genome and maps to human chromosome 5. A 4.3-kb DNA fragment of the mouse IL-13 gene (IL13) has been sequenced and found to occur as a single copy, mapping to mouse chromosome 11. Intrachromosomal mapping studies revealed that both genes contain four exons and three introns and show a high degree of sequence identity throughout their length. Potential recognition sequences for transcription factors that are present in the 5'-flanking region and are conserved between both genes include IFN-responsive elements, binding sites for AP-1, AP-2 and AP-3, and NF-IL 6 site, and a TATA-like sequence. Both genes map to chromosomal locations adjacent to genes encoding other cytokines, including IL-3, GM-CSF, IL-5, and IL-4, suggesting that IL-13 is another member of this cytokine gene family that may have arisen by gene duplication.

    View details for Web of Science ID A1993LH97800024

    View details for PubMedID 8099936

  • DINUCLEOTIDE REPEAT POLYMORPHISM AT THE HUMAN PITUITARY ADENYLATE-CYCLASE ACTIVATING POLYPEPTIDE (PACAP) GENE HUMAN MOLECULAR GENETICS PEREZJURADO, L. A., FRANCKE, U. 1993; 2 (6): 827-827

    View details for Web of Science ID A1993LH89200053

    View details for PubMedID 8353512

  • THE VIN-1 GENE, IDENTIFIED BY PROVIRUS INSERTIONAL MUTAGENESIS, IS THE CYCLIN D2 ONCOGENE Hanna, Z., Jankowski, M., Tremblay, P., Jiang, X. Y., MILATOVICH, A., FRANCKE, U., Jolicoeur, P. 1993; 8 (6): 1661-1666

    Abstract

    The Vin-1 gene was initially identified as a gene whose expression is altered by the integration of proviruses in the Vin-1 common site of integration in retrovirus-induced rodent T-cell leukemias. We have now isolated the Vin-1 cDNA. Sequencing of the Vin-1 cDNA and Vin-1 exons revealed that the proviruses are integrated at the 5' end of the Vin-1 gene in an inverse transcriptional orientation. The sequence of the Vin-1 gene is identical to that of the recently identified G1-phase cyclin D2 gene. The human homolog of the Vin-1/cyclin D2 gene (CCND2) was mapped to chromosome 12, band p13.3, by in situ hybridization, confirming previous mapping data. Our results strongly support a role of the cyclin D2 gene in oncogenesis and thereby implicate altered cell cycle regulation in transformation.

    View details for Web of Science ID A1993LE06400030

    View details for PubMedID 8502486

  • DIVERSE GROWTH-HORMONE RECEPTOR GENE-MUTATIONS IN LARON SYNDROME AMERICAN JOURNAL OF HUMAN GENETICS Berg, M. A., Argente, J., Chernausek, S., Gracia, R., GUEVARAAGUIRRE, J., Hopp, M., PEREZJURADO, L., Rosenbloom, A., Toledo, S. P., FRANCKE, U. 1993; 52 (5): 998-1005

    Abstract

    To better understand the molecular genetic basis and genetic epidemiology of Laron syndrome (growth-hormone insensitivity syndrome), we analyzed the growth-hormone receptor (GHR) genes of seven unrelated affected individuals from the United States, South America, Europe, and Africa. We amplified all nine GHR gene exons and splice junctions from these individuals by PCR and screened the products for mutations by using denaturing gradient gel electrophoresis (DGGE). We identified a single GHR gene fragment with abnormal DGGE results for each affected individual, sequenced this fragment, and, in each case, identified a mutation likely to cause Laron syndrome, including two nonsense mutations (R43X and R217X), two splice-junction mutations, (189-1 G to T and 71 + 1 G to A), and two frameshift mutations (46 del TT and 230 del TA or AT). Only one of these mutations, R43X, has been previously reported. Using haplotype analysis, we determined that this mutation, which involves a CpG dinucleotide hot spot, likely arose as a separate event in this case, relative to the two prior reports of R43X. Aside from R43X, the mutations we identified are unique to patients from particular geographic regions. Ten GHR gene mutations have now been described in this disorder. We conclude that Laron syndrome is caused by diverse GHR gene mutations, including deletions, RNA processing defects, translational stop codons, and missense codons. All the identified mutations involve the extracellular domain of the receptor, and most are unique to particular families or geographic areas.

    View details for Web of Science ID A1993LB88900018

    View details for PubMedID 8488849

  • STRUCTURE AND FUNCTIONAL EXPRESSION OF THE HUMAN MACROPHAGE INFLAMMATORY PROTEIN 1-ALPHA RANTES RECEPTOR JOURNAL OF EXPERIMENTAL MEDICINE Gao, J. L., Kuhns, D. B., Tiffany, H. L., McDermott, D., Li, X., FRANCKE, U., Murphy, P. M. 1993; 177 (5): 1421-1427

    Abstract

    The chemokine beta family is comprised of at least six distinct cytokines that regulate trafficking of phagocytes and lymphocytes in mammalian species; at least one of these, macrophage inflammatory protein 1 alpha (MIP-1 alpha), also regulates the growth of hematopoietic stem cells. We now show that MIP-1 alpha and the related beta chemokine, RANTES, induce transient alterations in intracellular Ca2+ concentration in polymorphonuclear leukocytes that can be reciprocally and specifically desensitized, suggesting a common receptor. Moreover, we have now cloned both the cDNA and the gene for this receptor, functionally expressed the receptor in Xenopus oocytes, and mapped the gene to human chromosome 3p21. Transcripts for the receptor were found in mature and immature myeloid cells as well as B cells. The receptor is a member of the G protein-coupled receptor superfamily. It has approximately 33% amino acid identity with receptors for the alpha chemokine, interleukin 8, and may be the human homologue of the product of US28, an open reading frame of human cytomegalovirus.

    View details for Web of Science ID A1993KY84900019

    View details for PubMedID 7683036

  • GENES FOR THE DIMERIZATION COFACTOR OF HEPATOCYTE NUCLEAR FACTOR-1-ALPHA (DCOH) ARE ON HUMAN AND MURINE CHROMOSOMES-10 GENOMICS MILATOVICH, A., Mendel, D. B., Crabtree, G. R., FRANCKE, U. 1993; 16 (1): 292-295

    Abstract

    Hepatocyte nuclear factor-1 alpha (HNF-1 alpha; gene symbol, TCF1) forms dimers with itself as well as with HNF-1 beta and regulates the expression of several liver-specific genes. Recently, a dimerization cofactor of hepatocyte nuclear factor-1 alpha, called DCOH, has been identified. Here, we report the chromosomal localization of the genes for this cofactor to chromosomes 10 in both humans and mice by Southern blot analyses of somatic cell hybrids.

    View details for Web of Science ID A1993KW20500050

    View details for PubMedID 8486378

  • DLX-1 AND DLX-2, 2 HOMEOBOX GENES EXPRESSED IN THE DEVELOPING FOREBRAIN, ARE CLOSELY LINKED ON MOUSE CHROMOSOME-2 MCGUINNESS, T. L., MACDONALD, G. P., FRANCKE, U., Ozcelik, T., Bulfone, A., Xu, D., Rubenstein, J. L. SLACK INC. 1993: A223–A223
  • MOLECULAR-CLONING, CDNA SEQUENCE, AND CHROMOSOMAL ASSIGNMENT OF THE HUMAN RADIXIN GENE AND 2 DISPERSED PSEUDOGENES GENOMICS Wilgenbus, K. K., MILATOVICH, A., FRANCKE, U., FURTHMAYR, H. 1993; 16 (1): 199-206

    Abstract

    Radixin is a cytoskeletal protein that may be important in linking actin to the plasma membrane. Recent cloning of the murine and porcine radixin cDNAs revealed a protein highly homologous to ezrin and moesin. We have cloned and sequenced the human radixin cDNA and found the predicted amino acid sequence for the human protein to be nearly identical to those predicted for radixin in the two other species. By Southern analyses of Chinese hamster x human somatic cell hybrid DNA and of PCR products derived from hybrids, the coding gene (RDX) was mapped to 11q. Fluorescence chromosomal in situ hybridization with a cDNA plasmid further localized this gene to band 11q23. However, PCR amplification with "radixin-specific" primers on the hybrid DNA panel yielded an additional, very similar DNA sequence that was further characterized by direct sequencing of PCR products. This sequence represents a truncated version and the respective locus (RDXP2) was assigned to Xp21.3. Furthermore, by employing a different set of primers, a third sequence was found that was 90% identical to the radixin sequence but contained termination codons and seemed to lack introns. This pseudogene (RDXP1) was mapped to 11p by Southern and PCR analyses.

    View details for Web of Science ID A1993KW20500027

    View details for PubMedID 8486357

  • A SERINE PROLINE-RICH PROTEIN IS FUSED TO HRX IN T(4-11) ACUTE LEUKEMIAS BLOOD Morrissey, J., Tkachuk, D. C., MILATOVICH, A., FRANCKE, U., Link, M., Cleary, M. L. 1993; 81 (5): 1124-1131

    Abstract

    Translocations involving chromosome band 11q23 in acute leukemias have recently been shown to involve the HRX gene that codes for a protein with significant similarity to Drosophila trithorax. HRX gene alterations are consistently observed in t(4;11) (q21;q23)-carrying leukemias and cell lines by Southern blot analyses and are accompanied by HRX transcripts of anomalous size on Northern blots. HRX-homologous cDNAs were isolated from a library prepared from t(4;11)-carrying acute leukemia cells. cDNAs representative of transcription products from the derivative 11 chromosome were shown to contain HRX sequences fused to sequences derived from chromosome band 4q21. Fragments of the latter were used to clone and analyze cDNAs for wild-type 4q21 transcripts that predicted a 140-Kd basic protein (named FEL) that is rich in prolines, serines, and charged amino acids. FEL contains guanosine triphosphate-binding and nuclear localization consensus sequences and uses one of two possible 5' exons encoding the first 12 or 5 amino acids. After t(4;11) translocations, 913 C-terminal amino acids of FEL are fused in frame to the N-terminal portion of HRX containing its minor groove DNA binding motifs. These features are similar to predicted t(11;19) fusion proteins, suggesting that HRX consistently contributes a novel DNA-binding motif to at least two different chimeric proteins in acute leukemias.

    View details for Web of Science ID A1993KP97700002

    View details for PubMedID 8443374

  • THE CD40 LIGAND, GP39, IS DEFECTIVE IN ACTIVATED T-CELLS FROM PATIENTS WITH X-LINKED HYPER-IGM SYNDROME CELL Aruffo, A., Farrington, M., Hollenbaugh, D., Li, X., MILATOVICH, A., Nonoyama, S., Bajorath, J., GROSMAIRE, L. S., Stenkamp, R., Neubauer, M., Roberts, R. L., Noelle, R. J., LEDBETTER, J. A., FRANCKE, U., Ochs, H. D. 1993; 72 (2): 291-300

    Abstract

    The prominent role of the CD40 receptor in B cell responses led us to investigate the role of the gp39-CD40 interaction in a group of primary immunodeficient patients with defective antibody production. Here we report that patients with hyper-IgM syndrome (HIM) have a defective gp39-CD40 interaction. B cells from HIM patients express functional CD40, but their T cells do not bind CD40-Ig. These patients expressed normal levels of gp39 mRNA, but these mRNAs encode defective gp39 proteins owing to mutations in the extracellular domain of gp39. Soluble recombinant forms of gp39 containing these mutations were unable to bind CD40 and drive normal B cell proliferation. The gene encoding gp39 was mapped to Xq26, the X chromosome region where the gene responsible for HIM had previously been mapped. These data suggest that a defect in gp39 is the basis of X-linked HIM.

    View details for Web of Science ID A1993KK03800013

    View details for PubMedID 7678782

  • Genes and gene products involved in Marfan syndrome. Seminars in thoracic and cardiovascular surgery FRANCKE, U., FURTHMAYR, H. 1993; 5 (1): 3-10

    View details for PubMedID 8425000

  • LARON SYNDROME (GROWTH-HORMONE RECEPTOR DEFICIENCY) IN ECUADOR - CLINICAL AND BIOCHEMICAL CHARACTERISTICS ARES-SERONO SYMP ON LESSONS FROM LARON SYNDROME ( LS ) 1966-1992 : A MODEL OF GH AND IGF-1 ACTION AND INTERACTION Rosenbloom, A. L., GUEVARAAGUIRRE, J., Fielder, P. J., GARGOSKY, S., Rosenfeld, R. G., Berg, M. A., FRANCKE, U., Diamond, F. B., VACCARELLO, M. A. KARGER. 1993: 34–52
  • LARON SYNDROME IN ECUADOR - POINT MUTATION IN THE GROWTH-HORMONE RECEPTOR GENE CAUSES DELETION OF 8 AMINO-ACIDS FROM THE EXTRACELLULAR DOMAIN ARES-SERONO SYMP ON LESSONS FROM LARON SYNDROME ( LS ) 1966-1992 : A MODEL OF GH AND IGF-1 ACTION AND INTERACTION Berg, M. A., GUEVARAAGUIRRE, J., Rosenbloom, A. L., Rosenfeld, R. G., FRANCKE, U. KARGER. 1993: 140-?
  • LARON-TYPE DWARFISM (LARON SYNDROME) IN ECUADOR 9th International Congress of Endocrinology: Progress in Endocrinology Rosenfeld, R. G., Rosenbloom, A. L., GUEVARAAGUIRRE, J., Berg, M. A., Cohen, P., Diamond, F. B., Fielder, P. J., FRANCKE, U., Gargosky, S. E., VACCARELLO, M. A., Wilson, K. PARTHENON PUBLISHING GROUP LTD. 1993: 265–267
  • Report of the first international workshop on human chromosome 18 mapping. Cytogenetics and cell genetics Le Beau, M. M., Overhauser, J., Straub, R. E., Silverman, G., Gilliam, T. C., Ott, J., O'Connell, P., FRANCKE, U., Geurts van Kessel, A. 1993; 63 (2): 78-96

    View details for PubMedID 8096808

  • MATERNAL IMPRINTING OF THE MOUSE SNRPN GENE AND CONSERVED LINKAGE HOMOLOGY WITH THE HUMAN PRADER-WILLI SYNDROME REGION NATURE GENETICS Leff, S. E., Brannan, C. I., Reed, M. L., Ozcelik, T., FRANCKE, U., Copeland, N. G., Jenkins, N. A. 1992; 2 (4): 259-264

    Abstract

    Prader-Willi syndrome (PWS) is associated with paternal gene deficiencies in human chromosome 15q11-13, suggesting that PWS is caused by a deficiency in one or more maternally imprinted genes. We have now mapped a gene, Snrpn, encoding a brain-enriched small nuclear ribonucleoprotein (snRNP)-associated polypeptide SmN, to mouse chromosome 7 in a region of homology with human chromosome 15q11-13 and demonstrated that Snrpn is a maternally imprinted gene in mouse. These studies, in combination with the accompanying human mapping studies showing that SNRPN maps in the Prader-Willi critical region, identify SNRPN as a candidate gene involved in PWS and suggest that PWS may be caused, in part, by defects in mRNA processing.

    View details for Web of Science ID A1992KB49500007

    View details for PubMedID 1303276

  • SMALL NUCLEAR RIBONUCLEOPROTEIN POLYPEPTIDE-N (SNRPN), AN EXPRESSED GENE IN THE PRADER-WILLI SYNDROME CRITICAL REGION NATURE GENETICS Ozcelik, T., Leff, S., Robinson, W., Donlon, T., Lalande, M., SANJINES, E., Schinzel, A., FRANCKE, U. 1992; 2 (4): 265-269

    Abstract

    Prader-Willi syndrome (PWS) is associated with paternally derived chromosomal deletions in region 15q11-13 or with maternal disomy for chromosome 15. Therefore, loss of the expressed paternal alleles of maternally imprinted genes must be responsible for the PWS phenotype. We have mapped the gene encoding the small nuclear RNA associated polypeptide SmN (SNRPN) to human chromosome 15q12 and a processed pseudogene SNRPNP1 to chromosome region 6pter-p21. Furthermore, SNRPN was mapped to the minimal deletion interval that is critical for PWS. The fact that the mouse Snrpn gene is maternally imprinted in brain suggests that loss of the paternally derived SNRPN allele may be involved in the PWS phenotype.

    View details for Web of Science ID A1992KB49500008

    View details for PubMedID 1303277

  • Serotonin receptor 1c gene assigned to X chromosome in human (band q24) and mouse (bands D-F4). Human molecular genetics MILATOVICH, A., Hsieh, C. L., Bonaminio, G., Tecott, L., Julius, D., FRANCKE, U. 1992; 1 (9): 681-684

    Abstract

    In the mammalian nervous system, serotonin (5-hydroxytryptamine) binds to distinct cell surface receptor subtypes that are defined by their ligand binding and effector-coupling properties. The 5HT1c receptor is a G-protein coupled receptor that stimulates phospholipase C-catalyzed hydrolysis of phosphatidylinositol bisphosphate, leading to the mobilization of intracellular calcium and to the activation of protein kinase C. By using somatic cell hybrid analysis and FISH, we have mapped the HTR1C locus to the human X chromosome, band q24 and to the mouse X chromosome region D-F4. Comparison of these map positions offers new insights into the evolution of human and murine X chromosomes. Since HTR1C is expressed in certain parts of the central nervous system and abnormal function of the serotoninergic system has been implicated in affective disorders, obsessive-compulsive disorder and epilepsy, establishing the precise map position of HTR1C is an important first step toward evaluating this locus as a candidate for mutations in these syndromes and in X-linked mental disorders.

    View details for PubMedID 1302605

  • AN X-LINKED HUMAN COLLAGEN TRANSGENE ESCAPES X-INACTIVATION IN A SUBSET OF CELLS DEVELOPMENT Wu, H., Fassler, R., Schnieke, A., Barker, D., Lee, K. H., Chapman, V., FRANCKE, U., Jaenisch, R. 1992; 116 (3): 687-695

    Abstract

    Transgenic mice carrying one complete copy of the human alpha 1(I) collagen gene on the X chromosome (HucII mice) were used to study the effect of X inactivation on transgene expression. By chromosomal in situ hybridization, the transgene was mapped to the D/E region close to the Xce locus, which is the controlling element. Quantitative RNA analyses indicated that transgene expression in homozygous and heterozygous females was about 125% and 62%, respectively, of the level found in hemizygous males. Also, females with Searle's translocation carrying the transgene on the inactive X chromosome (Xi) expressed about 18% transgene RNA when compared to hemizygous males. These results were consistent with the transgene being subject to but partially escaping from X inactivation. Two lines of evidence indicated that the transgene escaped X inactivation or was reactivated in a small subset of cells rather than being expressed at a lower level from the Xi in all cells, (i) None of nine single cell clones carrying the transgene on the Xi transcribed transgene RNA. In these clones the transgene was highly methylated in contrast to clones carrying the transgene on the Xa. (ii) In situ hybridization to RNA of cultured cells revealed that about 3% of uncloned cells with the transgene on the Xi expressed transgene RNA at a level comparable to that on the Xa. Our results indicate that the autosomal human collagen gene integrated on the mouse X chromosome is susceptible to X inactivation. Inactivation is, however, not complete as a subset of cells carrying the transgene on Xi expresses the transgene at a level comparable to that when carried on Xa.

    View details for Web of Science ID A1992KB02500016

    View details for PubMedID 1289060

  • LOCALIZATION OF THE PHOTORECEPTOR GENE ROMI TO HUMAN CHROMOSOME-11 AND MOUSE CHROMOSOME-19 - SUBLOCALIZATION TO HUMAN 11Q13 BETWEEN PGA AND PYGM AMERICAN JOURNAL OF HUMAN GENETICS Bascom, R. A., GARCIAHERAS, J., Hsieh, C. L., Gerhard, D. S., Jones, C., FRANCKE, U., WILLARD, H. F., Ledbetter, D. H., McInnes, R. R. 1992; 51 (5): 1028-1035

    Abstract

    Rom-1 is a retinal integral membrane protein that, together with the product of the human retinal degeneration slow gene (RDS), defines a photoreceptor-specific protein family. The gene for rom-1 (HGM symbol: ROM1) has been assigned to human chromosome 11 and mouse chromosome 19 by Southern blot analysis of somatic cell hybrid DNAs. ROM1 was regionally sublocalized to human 11p13-11q13 by using three mouse-human somatic cell hybrids; in situ hybridization refined the sublocalization to human 11q13. Analysis of somatic cell hybrids suggested that the most likely localization of ROM1 is in the approximately 2-cM interval between human PGA (human pepsinogen A) and PYGM (muscle glycogen phosphorylase). ROM1 appears to be a new member of a conserved syntenic group whose members include such genes as CD5, CD20, and OSBP (oxysterol-binding protein), on human chromosome 11 and mouse chromosome 19. Localization of the ROM1 gene will permit the examination of its linkage to hereditary retinopathies in man and mouse.

    View details for Web of Science ID A1992JU84000012

    View details for PubMedID 1415249

  • TISSUE-SPECIFIC EXPRESSION AND CHROMOSOME ASSIGNMENT OF GENES SPECIFYING 2 ISOFORMS OF SUBUNIT-VIIA OF HUMAN CYTOCHROME-C-OXIDASE GENE Arnaudo, E., Hirano, M., Seelan, R. S., MILATOVICH, A., Hsieh, C. L., Fabrizi, G. M., Grossman, L. I., FRANCKE, U., Schon, E. A. 1992; 119 (2): 299-305

    Abstract

    Subunit VIIa of mammalian cytochrome c oxidase (COX; EC 1.9.3.1) exists in at least two isoforms, one present in all tissue types ('liver' isoform; COX VIIa-L) and the other specific for cardiac and skeletal muscle (COX VIIa-M). We have isolated a full-length cDNA encoding human COX VIIa-M. The deduced polypeptide represents the human ortholog of COX VIIa-M, as it shares 78% identity with bovine COX VIIa-M, but only 63% identity with human COX VIIa-L. Northern-blot analysis of primate tissues demonstrated that COXVIIa-M mRNA is present only in muscle tissues; in contrast, the COXVIIa-L mRNA is present in both muscle and nonmuscle tissues. Southern-blot hybridization of human-rodent cell hybrid genomic DNA indicates that the COXVIIa-M gene maps to a single locus on chromosome 19, designated COX7AM. In contrast, COXVIIa-L cDNA probes hybridized to fragments from two COX7AL loci, on chromosomes 4 and 14.

    View details for Web of Science ID A1992JQ15300021

    View details for PubMedID 1327965

  • MOLECULAR EVOLUTION OF THE HUMAN INTERLEUKIN-8 RECEPTOR GENE-CLUSTER NATURE GENETICS Ahuja, S. K., Ozcelik, T., MILATOVITCH, A., FRANCKE, U., Murphy, P. M. 1992; 2 (1): 31-36

    Abstract

    Interleukin-8 (IL-8) is the prototype for a family of at least eight neutrophil chemoattractants whose genes map to human chromosome 4q13-q21. Two human IL-8 receptors, IL8RA and IL8RB, are known from cDNA cloning; IL8RA is a promiscuous receptor for at least two other related ligands, GRO alpha and NAP-2. We now report cloning of the genes for IL8RA, IL8RB and a recently inactivated pseudogene of receptor A (IL8RAP). These form a cluster of only three genes in the superfamily of G protein-coupled receptors (GPCRs) and map to 2q34-q35. The coevolutionary diversity displayed by the IL-8 ligand-receptor complex--ligand promiscuity for IL-8, receptor promiscuity for IL8RA, gene duplication for both ligands and receptors and gene extinction in the case of IL8RAP--is unprecedented for the GPCR superfamily.

    View details for Web of Science ID A1992JM64600012

    View details for PubMedID 1303245

  • STRUCTURE AND CHROMOSOMAL LOCALIZATION OF THE MAMMALIAN AGRIN GENE JOURNAL OF NEUROSCIENCE Rupp, F., Ozcelik, T., Linial, M., Peterson, K., FRANCKE, U., Scheller, R. 1992; 12 (9): 3535-3544

    Abstract

    Agrin, a component of the synaptic basal lamina, has been shown to induce clustering of ACh receptors on the surface of muscle fibers. Analysis of cDNAs isolated from a rat embryonic spinal cord library demonstrated that agrin contains domains similar to regions of protease inhibitors, laminin and epidermal growth factor. The domain structure of agrin is further revealed here in an analysis of the agrin gene. Two additional internal repeated sequences are defined: one rich in cysteine residues with no homology to other proteins, and another similar to the laminin G domain, which is involved in heparin binding. Alternative RNA splicing at two positions in the gene predicts up to eight possible forms of the agrin protein. The gene (symbol AGRN/Agrn) has been assigned to chromosome 1 region pter-p32 in human and to mouse chromosome 4.

    View details for Web of Science ID A1992JN80700021

    View details for PubMedID 1326608

  • MOLECULAR-GENETICS OF STEROID 5-ALPHA-REDUCTASE-2 DEFICIENCY JOURNAL OF CLINICAL INVESTIGATION Thigpen, A. E., Davis, D. L., MILATOVICH, A., Mendonca, B. B., IMPERATOMCGINLEY, J., Griffin, J. E., FRANCKE, U., Wilson, J. D., Russell, D. W. 1992; 90 (3): 799-809

    Abstract

    Two isozymes of steroid 5 alpha-reductase encoded by separate loci catalyze the conversion of testosterone to dihydrotestosterone. Inherited defects in the type 2 isozyme lead to male pseudohermaphroditism in which affected males have a normal internal urogenital tract but external genitalia resembling those of a female. The 5 alpha-reductase type 2 gene (gene symbol SRD5A2) was cloned and shown to contain five exons and four introns. The gene was localized to chromosome 2 band p23 by somatic cell hybrid mapping and chromosomal in situ hybridization. Molecular analysis of the SRD5A2 gene resulted in the identification of 18 mutations in 11 homozygotes, 6 compound heterozygotes, and 4 inferred compound heterozygotes from 23 families with 5 alpha-reductase deficiency. 6 apparent recurrent mutations were detected in 19 different ethnic backgrounds. In two patients, the catalytic efficiency of the mutant enzymes correlated with the severity of the disease. The high proportion of compound heterozygotes suggests that the carrier frequency of mutations in the 5 alpha-reductase type 2 gene may be higher than previously thought.

    View details for Web of Science ID A1992JN95900017

    View details for PubMedID 1522235

  • DLX2 (TES1), A HOMEOBOX GENE OF THE DISTAL-LESS FAMILY, ASSIGNED TO CONSERVED REGIONS ON HUMAN AND MOUSE CHROMOSOMES-2 GENOMICS Ozcelik, T., Porteus, M. H., Rubenstein, J. L., FRANCKE, U. 1992; 13 (4): 1157-1161

    Abstract

    Dlx-2 (also called Tes-1), a mammalian member of the Distal-less family of homeobox genes, is expressed during murine fetal development in spatially restricted domains of the forebrain. Searching for a candidate neurological mutation that might involve this gene, we have assigned the human and mouse loci to regions of conserved synteny on human chromosome 2, region cen--q33, and mouse chromosome 2 by Southern analysis of somatic cell hybrid lines. An EcoRI dimorphism, discovered in common inbred laboratory strains, was used for recombinant inbred strain mapping. The results place Dlx-2/Tes-1 near the Hox-4 cluster on mouse chromosome 2.

    View details for Web of Science ID A1992JH14800031

    View details for PubMedID 1354641

  • Common sequence motifs at the rearrangement sites of a constitutional X/autosome translocation and associated deletion. American journal of human genetics GIACALONE, J. P., FRANCKE, U. 1992; 51 (1): 226-?

    View details for PubMedID 1609802

  • THE GENE FOR THE PERIPHERAL MYELIN PROTEIN-PMP-22 IS A CANDIDATE FOR CHARCOT-MARIE-TOOTH DISEASE TYPE-1A NATURE GENETICS Patel, P. I., Roa, B. B., Welcher, A. A., SCHOENERSCOTT, R., Trask, B. J., Pentao, L., Snipes, G. J., Garcia, C. A., FRANCKE, U., Shooter, E. M., Lupski, J. R., Suter, U. 1992; 1 (3): 159-165

    Abstract

    Charcot-Marie-Tooth disease type 1A (CMT1A) is an autosomal dominant peripheral neuropathy associated with a large DNA duplication on the short arm of human chromosome 17. The trembler (Tr) mouse serves as a model for CMT1A because of phenotypic similarities and because the Tr locus maps to mouse chromosome 11 in a region of conserved synteny with human chromosome 17. Recently, the peripheral myelin gene Pmp-22 was found to carry a point mutation in Tr mice. We have isolated cDNA and genomic clones for human PMP-22. The gene maps to human chromosome 17p11.2-17p12, is expressed at high levels in peripheral nervous tissue and is duplicated, but not disrupted, in CMT1A patients. Thus, we suggest that a gene dosage effect involving PMP-22 is at least partially responsible for the demyelinating neuropathy seen in CMT1A.

    View details for Web of Science ID A1992JC98200005

    View details for PubMedID 1303228

  • RECOMBINATION OF 4P16 DNA MARKERS IN AN UNUSUAL FAMILY WITH HUNTINGTON DISEASE AMERICAN JOURNAL OF HUMAN GENETICS Pritchard, C., Zhu, N., Zuo, J., Bull, L., PERICAKVANCE, M. A., Vance, J. M., Roses, A. D., MILATOVICH, A., FRANCKE, U., Cox, D. R., Myers, R. M. 1992; 50 (6): 1218-1230

    Abstract

    The Huntington disease (HD) mutation has been localized to human chromosome 4p16, in a 6-Mb region between the D4S10 locus and the 4p telomere. In a report by Robbins et al., a family was identified in which an affected individual failed to inherit three alleles within the 6-Mb region originating from the parental HD chromosome. To explain these results, it was suggested that the HD locus (HD) lies close to the telomere and that a recombination event took place between HD and the most telomeric marker examined, D4S90. As a test of this telomere hypothesis, we examined six members of this family, five of whom are affected with HD, for the segregation of 12 polymorphic markers from 4p16, including D4S169, which lies within 80 kb of the 4p telomere. We separated, in somatic cell hybrids, the chromosomes 4 from each family member, to determine the phase of marker alleles on each chromosome. We excluded nonpaternity by performing DNA fingerprint analyses on all six family members, and we found no evidence for chromosomal rearrangements when we used high-resolution karyotype analysis. We found that two affected siblings, including one of the patients originally described by Robbins et al., inherited alleles from the non-HD chromosome 4 of their affected parents, throughout the 6-Mb region. We found that a third affected sibling, also studied by Robbins et al., inherited alleles from the HD chromosome 4 of the affected parent, throughout the 6-Mb region. Finally, we found that a fourth sibling, who is likely affected with HD, has both a recombination event within the 6-Mb region and an additional recombination event in a more centromeric region of the short arm of chromosome 4. Our results argue against a telomeric location for HD and suggest that the HD mutation in this family is either associated with DNA predisposed to double recombination and/or gene conversion within the 6-Mb region or is in a gene that is outside this region and that is different from that mutated in most other families with HD.

    View details for Web of Science ID A1992HY65400009

    View details for PubMedID 1350884

  • MUTATION ANALYSIS AND PRENATAL-DIAGNOSIS IN A LESCH-NYHAN FAMILY SHOWING NONRANDOM X-INACTIVATION INTERFERING WITH CARRIER DETECTION TESTS HUMAN GENETICS Marcus, S., Steen, A. M., ANDERSSON, B., Lambert, B., Kristoffersson, U., FRANCKE, U. 1992; 89 (4): 395-400

    Abstract

    A nonsense mutation at the CpG-site in the codon for Arg(169) in the gene for hypoxanthine phosphoribosyltransferase (hprt) was identified by genomic polymerase chain reaction (PCR) and DNA sequencing in cultured fibroblasts from two brothers with Lesch Nyhan's syndrome. The recurrence of mutation at this CpG-site in several unrelated Lesch-Nyhan families suggests that deamination of 5-methylcytosine is a possible mechanism for mutagenesis. The level of hprt-mRNA in the fibroblasts of the patients was similar to that in healthy controls, whereas hprt-enzyme activity was not detectable. The mutation in this family was also identified in five female relatives and prenatally in a male fetus. Unexpectedly, results from hair follicle analyses and fibroblast selection studies in 8-azaguanine and 6-thioguanine medium showed a non-carrier phenotype in three of the female heterozygotes, whereas X-inactivation mosaicism was demonstrated in one heterozygote. A possible explanation for the apparent non-random X-inactivation in this family is the co-existence of the hprt mutation with an undefined X-linked lethal mutation. This observation is of practical relevance for carrier detection in other Lesch-Nyhan families.

    View details for Web of Science ID A1992JA74200006

    View details for PubMedID 1618489

  • HUMAN CYCLIN-B1 GENE (CCNB1) ASSIGNED TO CHROMOSOME-5 (Q13-QTER) SOMATIC CELL AND MOLECULAR GENETICS MILATOVICH, A., FRANCKE, U. 1992; 18 (3): 303-307

    Abstract

    Cyclins play an important role in cell cycle regulation. At least five classes of cyclins have been identified--A, B, C, D, and E. B cyclins are generally of two types in most organisms--B1 and B2. We have mapped the gene for human cyclin B1 (CCNB1) to human chromosome 5 (region q13-qter) by Southern blot analysis of human x Chinese hamster somatic cell hybrid panels. Many more cyclin B-related sequences have been identified in the mouse (Cycb-1 to Cycb-10) and have been mapped to chromosomes 4, 5, 7, 8, 13, 14, 15, and 17. Based on our mapping of human CCNB1 and known evolutionary conservation of chromosomal regions, we propose that the homologous cyclin B1 locus, Cycb-4, on mouse chromosome 13 is a functional gene.

    View details for Web of Science ID A1992JH56100009

    View details for PubMedID 1386686

  • ESTIMATION OF THE MALE AND FEMALE MUTATION-RATES IN DUCHENNE MUSCULAR-DYSTROPHY (DMD) HUMAN GENETICS Muller, B., DeChant, C., Meng, G., LIECHTIGALLATI, S., DOHERTY, R. A., HEITMANCIK, J. F., Bakker, E., Read, A. P., JEANPIERRE, M., FISCHBECK, K. H., Romeo, G., FRANCKE, U., Wilichowski, E., GREENBERG, C. R., Van Broeckhoven, C., JUNIEN, C., Muller, C. R., Grimm, T. 1992; 89 (2): 204-206

    Abstract

    We present the results of an international collaborative study aimed at estimating the ratio of male to female mutation rates in Duchenne muscular dystrophy based on the method of C. Müller and T. Grimm. With a sample size of 295, this ratio is found to be very close to 1, thus giving evidence for equal mutation rates in males and females in Duchenne muscular dystrophy.

    View details for Web of Science ID A1992HU97900012

    View details for PubMedID 1587532

  • PHENOTYPIC, CYTOGENETIC, AND MOLECULAR STUDIES OF 3 PATIENTS WITH CONSTITUTIONAL DELETIONS OF CHROMOSOME-5 IN THE REGION OF THE GENE FOR FAMILIAL ADENOMATOUS POLYPOSIS AMERICAN JOURNAL OF HUMAN GENETICS Lindgren, V., Bryke, C. R., Ozcelik, T., YANGFENG, T. L., FRANCKE, U. 1992; 50 (5): 988-997

    Abstract

    We have studied three patients, one with extensive polyposis of the colon, who have constitutional interstitial deletions of the long arm of chromosome 5. High-resolution banding studies indicated that the deletion in the patient with polyposis spans the region 5q21-q22, which includes APC, a gene involved in familial adenomatous polyposis and sporadic colon cancer. Molecular analysis with probes for sequences flanking APC confirmed this conclusion. The deletions in the other two patients, who are too young to have developed polyposis, had breakpoints within this region, precluding the use of cytogenetic analysis alone in making definitive predictions about their risks. Molecular studies resolved the uncertainty; in situ and quantitative Southern hybridizations of four probes for polymorphic segments revealed that one of the patients has a deletion of MCC, a gene which is approximately 150 kb proximal to APC, and two flanking markers. He is at increased risk for polyposis, while the other patient is not. The physical descriptions of these patients, in conjunction with cases in the literature, begin to allow delineation of two distinct 5q-syndromes. These studies also provide precise physical mapping data for D5S71, D5S81, D5S84, and MCC on 5q.

    View details for Web of Science ID A1992HR47300012

    View details for PubMedID 1315124

  • ASSIGNMENT OF THE HUMAN ALPHA-2-PLASMIN INHIBITOR GENE (PLI) TO CHROMOSOME 17, REGION PTER-P12, BY PCR ANALYSIS OF SOMATIC-CELL HYBRIDS GENOMICS Welch, S. K., FRANCKE, U. 1992; 13 (1): 213-214

    Abstract

    The human gene for the alpha 2-plasmin inhibitor (PLI) had been assigned by others to the pericentromeric region of chromosome 18 by in situ hybridization. However, when we used a probe for this gene in our efforts to construct a complete physical map of chromosome 18, we discovered that PLI could be excluded from this chromosome. On the basis of the published PLI sequence, we designed primers to sequences in intron 6 and 7 that direct amplification of a 353-bp fragment that includes the entire exon 7. By using PCR analysis of rodent x human hybrid panels, we have unequivocally assigned the PLI locus to human chromosome 17. With a regional mapping panel, the assignment could be narrowed to region 17pter-p12.

    View details for Web of Science ID A1992HM69900032

    View details for PubMedID 1577485

  • A NOVEL GC-RICH HUMAN MACROSATELLITE VNTR IN XQ24 IS DIFFERENTIALLY METHYLATED ON ACTIVE AND INACTIVE X-CHROMOSOMES NATURE GENETICS Giacalone, J., FRIEDES, J., FRANCKE, U. 1992; 1 (2): 137-143

    Abstract

    A new X chromosome-specific repetitive sequence, a 3 kilobase HindIII clone with a base composition of 63% C+G, has been isolated. The sequence is organized as a hypervariable tandem repeat cluster ranging in size from 150-350 kilobases, with outlying single copies. This locus, designated DXZ4 and mapped to chromosome band Xq24, may consist of as many as 50 variable-length alleles. It represents a class of variable number of tandem repeat polymorphism which may be termed 'macrosatellite'. The cluster is highly methylated on the active X chromosome and hypomethylated on the inactive X.

    View details for Web of Science ID A1992JC98000013

    View details for PubMedID 1302007

  • HUMAN CHROMOSOME-19 CONTAINS THE NEUROTROPHIN-5 GENE LOCUS AND 3 RELATED GENES THAT MAY ENCODE NOVEL ACIDIC NEUROTROPHINS SOMATIC CELL AND MOLECULAR GENETICS BERKEMEIER, L. R., Ozcelik, T., FRANCKE, U., Rosenthal, A. 1992; 18 (3): 233-245

    Abstract

    Differentiation, survival, and function of the vertebrate neurons are controlled by multiple, target-derived neurotrophic factors. The best characterized mammalian neurotrophic factors are four structurally related 13 to 14 kDa basic proteins, collectively known as neurotrophins. Here we describe the identification of a gene cluster localized on human chromosome 19 that contains neurotrophin-5 (NT-5) and that may encode three additional acidic members of this protein family. The three novel partial open reading frames (ORFs), designated neurotrophin-6-alpha (NT6-alpha), NT6-beta and NT6-gamma, are 95% identical to each other and 75% identical to NT5. The putative mature N-terminal portion of NT6 ORFs does not contain a typical dibasic cleavage site and lacks two out of six cysteines that are conserved among the neutrophins. The unique structures of NT6-alpha, -beta, and -gamma suggest that if the NT6 open reading frames indeed code for functional proteins, these proteins may display novel functions and may act through a distinct class of receptors. In the human, both NTF5 and NTF6 gene loci were mapped to chromosome 19 by Southern analysis of somatic cell hybrid panels. In mouse, the NT5 gene (Ntf-5) was assigned to chromosome 7 and no sequences representing NT6 homologs were identified.

    View details for Web of Science ID A1992JH56100004

    View details for PubMedID 1496419

  • A STRUCTURAL HOMOLOG OF THE N-FORMYL PEPTIDE RECEPTOR - CHARACTERIZATION AND CHROMOSOME MAPPING OF A PEPTIDE CHEMOATTRACTANT RECEPTOR FAMILY JOURNAL OF BIOLOGICAL CHEMISTRY Murphy, P. M., Ozcelik, T., Kenney, R. T., Tiffany, H. L., McDermott, D., FRANCKE, U. 1992; 267 (11): 7637-7643

    Abstract

    Phagocytic cells of many higher species express calcium mobilizing G protein-coupled receptors for bacterial N-formyl peptides which mediate chemotaxis, degranulation, and the respiratory burst. cDNA encoding an N-formyl peptide receptor (FPR) has been reported. We now report the isolation of a closely related cDNA, 2.6 kilobase pairs in length, which we have designated as the FPRL1 receptor cDNA (FPRL1 = formyl peptide receptor like-1). FPR and the FPRL1 receptor derive from small, single-copy genes, both of which are located on human chromosome 19. The gene loci are designated FPR1 and FPRL1, respectively. Both FPR and FPRL1 cDNA cross-hybridize under high stringency conditions with a third gene, designated as FPRL2, which does not appear to be expressed in neutrophils. In contrast, transcripts for both the FPRL1 receptor and FPR are detected only in differentiated myeloid cells; the distribution of N-formyl peptide binding sites is also restricted to mature myeloid cells. FPRL1 cDNA encodes a 351-amino acid polypeptide whose sequence is 69% identical to that of FPR. G protein-coupled receptors that exhibit this degree of structural similarity typically possess a conserved ligand specificity. However, the FPRL1 receptor does not bind prototype N-formyl peptide ligands when expressed in heterologous cell types. These results suggest that FPR1 may be the only gene that is expressed by neutrophils that encodes a receptor capable of binding prototype N-formyl peptides. Moreover, discovery of the FPRL1 receptor indicates the existence of another as yet unidentified peptide that may recruit neutrophils to sites of inflammation.

    View details for Web of Science ID A1992HN48500063

    View details for PubMedID 1373134

  • TREMBLER MOUSE CARRIES A POINT MUTATION IN A MYELIN GENE NATURE Suter, U., Welcher, A. A., Ozcelik, T., Snipes, G. J., Kosaras, B., FRANCKE, U., BILLINGSGAGLIARDI, S., Sidman, R. L., Shooter, E. M. 1992; 356 (6366): 241-244

    Abstract

    The autosomal dominant trembler mutation (Tr), maps to mouse chromosome 11 (ref. 2) and manifests as a Schwann-cell defect characterized by severe hypomyelination and continuing Schwann-cell proliferation throughout life. Affected animals move clumsily and develop tremor and transient seizures at a young age. We have recently described a potentially growth-regulating myelin protein, peripheral myelin protein-22 (PMP-22; refs 7, 8), which is expressed by Schwann cells and found in peripheral myelin. We now report the assignment of the gene for PMP-22 to mouse chromosome 11. Cloning and sequencing of PMP-22 complementary DNAs from inbred Tr mice reveals a point mutation that substitutes an aspartic acid residue for a glycine in a putative membrane-associated domain of the PMP-22 protein. Our results identify the PMP-22 gene as a likely candidate for the mouse trembler locus and will encourage the search for mutations in the corresponding human gene in pedigrees with hypertrophic neuropathies such as Charcot-Marie-Tooth and Dejerine-Sottas diseases (hereditary motor and sensory neuropathies I and III).

    View details for Web of Science ID A1992HJ94400058

    View details for PubMedID 1552943

  • CDNA CLONING OF A LIVER ISOFORM OF THE PHOSPHORYLASE-KINASE ALPHA-SUBUNIT AND MAPPING OF THE GENE TO XP22.2-P22.1, THE REGION OF HUMAN X-LINKED LIVER GLYCOGENOSIS PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Davidson, J. J., Ozcelik, T., Hamacher, C., Willems, P. J., FRANCKE, U., Kilimann, M. W. 1992; 89 (6): 2096-2100

    Abstract

    We have cloned cDNA molecules encoding another isoform of the alpha subunit of phosphorylase kinase (ATP:phosphorylase-b phosphotransferase, EC 2.7.1.38). Sequence comparison with the previously characterized muscle isoform reveals a pattern of highly conserved and variable domains and demonstrates that the isoforms are the products of distinct genes. In contrast to the muscle isoform gene, PHKA1, the gene of this additional isoform, PHKA2, is predominantly expressed in liver and other nonmuscle tissues. It was mapped to the distal short arm of the human X chromosome (Xp22.2-p22.1), the same region to which human X-linked liver glycogenosis due to phosphorylase kinase deficiency has been mapped. Thus, X-linked liver glycogenosis is probably caused by mutations affecting PHKA2.

    View details for Web of Science ID A1992HJ05300021

    View details for PubMedID 1372435

  • GROWTH-HORMONE RECEPTOR GENE MUTATION AND ABNORMAL RNA SPLICING IN ECUADORIAN PATIENTS WITH LARON SYNDROME Berg, M. A., Aguirre, J. G., Rosenbloom, A. L., Rosenfeld, R. G., FRANCKE, U. SLACK INC. 1992: A10–A10
  • PARTIAL DELETION OF A DYSTROPHIN GENE LEADS TO EXON SKIPPING AND TO LOSS OF AN INTRA-EXON HAIRPIN STRUCTURE FROM THE PREDICTED MESSENGER-RNA PRECURSOR BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS Matsuo, M., Nishio, H., Kitoh, Y., FRANCKE, U., Nakamura, H. 1992; 182 (2): 495-500

    Abstract

    In dystrophin Kobe exon 19 of the dystrophin gene is skipped during the process of mRNA precursor splicing even though the splice sites are unchanged (Matsuo et al. J. Clin. Invest. 87:2127-2131,1991). In the predicted secondary structure of the mRNA precursor, exon 19 of dystrophin Kobe is paired with intron sequences, whereas a large part of exon sequence from wild type is paired with itself and folded into a large hairpin structure. As all of 22 additional dystrophin exons analyzed also form intra-exon hairpin structures, these structures may be considered essential components of exons. We suggest that the abolishment of a hairpin structure in the truncated exon of dystrophin Kobe might prevent the splicing machinery from recognizing the splice sites and induce exon skipping.

    View details for Web of Science ID A1992HC39700010

    View details for PubMedID 1734864

  • INDUCED RECIPROCAL TRANSLOCATION IN TRANSGENIC MICE NEAR SITES OF TRANSGENE INTEGRATION MAMMALIAN GENOME FRANCKE, U., Hsieh, C. L., Kelly, D., Lai, E., Popko, B. 1992; 3 (4): 209-216

    Abstract

    Transgenic mice (JCP0 #18), heterozygous for an insertion of approximately 50 copies of the rat peripheral myelin (P0) protein cDNA, displayed a pattern of reduced litter size that suggested a chromosome rearrangement. Chromosome banding studies of fetal cells disclosed the presence of an apparently balanced translocation between a Chromosome (Chr) 1 and 14 with breakpoints at bands 1H3 and 14C3. In situ hybridization of biotin-labeled P0 rat cDNA probe to chromosome spreads and detection of specific signal with fluorescein isothiocyanate-conjugated avidin revealed a strong signal on the 1(14) translocation chromosome at the site of the breakpoint. A weaker signal was present near the breakpoint on the 14(1) derivative chromosome. These results suggest an etiologic relationship between the insertion of the transgene and the origin of the translocation. To further elucidate possible mechanisms, we first mapped the endogenous P0 gene (gene symbol Mpp). As previously reported (You et al., Genomics 9: 751, 1991), we found that Mpp is located on Chr 1 in the region of the translocation breakpoint in JCP0 mice. Subsequently, we have carried out pulsed-field gel and standard Southern analyses with P0 gene probes, but found no evidence for a direct involvement of the endogenous P0 gene in the process that generated the balanced reciprocal translocation. Thus, we favor the hypothesis that, during repair of DNA strand breakage--possibly induced by the microinjection procedure--the transgene copies were ligated to broken ends of Chrs 1 and 14. According to convention, this translocation is designated T(1;14)1Po. Homozygotes are phenotypically normal and breed well; they will be useful for genetic and physical mapping of Chrs 1 and 14.

    View details for Web of Science ID A1992JV42300005

    View details for PubMedID 1377063

  • Mutation creating a new splice site in the growth hormone receptor genes of 37 Ecuadorean patients with Laron syndrome. Human mutation Berg, M. A., Guevara-Aguirre, J., Rosenbloom, A. L., Rosenfeld, R. G., FRANCKE, U. 1992; 1 (1): 24-32

    Abstract

    Laron syndrome is an autosomal recessive condition characterized by resistance to growth hormone. We sought to determine the molecular basis of this condition in an Ecuadorean population with a high incidence of affected individuals. Growth hormone receptor gene sequences from an obligate heterozygote were amplified by the polymerase chain reaction and screened for mutations using denaturing gradient gel electrophoresis. Only exon 6 revealed homo- and heteroduplexes on denaturing gradient gels. Sequencing revealed a substitution of guanine for adenine in the third position of codon 180 that did not change the amino acid encoded. Sequencing of the exon 6-exon 7 splice junction from RNA-polymerase chain reaction amplified cellular RNA of an affected individual revealed that the substitution activates a 5' splice site 24 nucleotides upstream from the normal exon 6-intron 6 boundary. Splicing in two probands' lymphoblasts occurred virtually exclusively at the abnormal 5' splice site created by the codon 180 substitution. Exon 6 sequences from 38 patients and 47 relatives were amplified and analyzed by sequencing or dot-blot hybridization with allele-specific oligonucleotides. The substitution was detected in 74 of 76 Laron syndrome patients' GH-receptor alleles. All 26 parents and 12 of 21 unaffected siblings were heterozygous for this mutation. It was absent in 61 unrelated unaffected control individuals. We conclude that the codon 180 nucleotide substitution probably causes Laron syndrome as translation of the observed, abnormally spliced growth hormone receptor transcript would lead to the synthesis of a receptor protein with an 8 amino acid deletion from the extracellular domain.

    View details for PubMedID 1284474

  • GENE FOR LYMPHOID ENHANCER-BINDING FACTOR-I (LEF1) MAPPED TO HUMAN CHROMOSOME-4 (Q23-Q25) AND MOUSE CHROMOSOME-3 NEAR EGF GENOMICS MILATOVICH, A., Travis, A., Grosschedl, R., FRANCKE, U. 1991; 11 (4): 1040-1048

    Abstract

    LEF-1 is a 54-kDa nuclear protein that is expressed specifically in pre-B and T-cells. It binds to a functionally important site in the T-cell receptor alpha enhancer and contributes to maximal enhancer activity. LEF-1 is a member of a family of regulatory proteins that share homology with the high mobility group protein 1 (HMG1). The location of the LEF1 gene on human and mouse chromosomes was determined by Southern blot analysis of DNA from panels of interspecies somatic cell hybrids using a murine cDNA probe. Human-specific DNA fragments were detected in all somatic cell hybrids that retained the human chromosomal region 4cen-q31.2. Fluorescent in situ hybridization with two biotin-labeled overlapping human genomic cosmids revealed a specific hybridization signal at 4q23-q25. The homologous locus in the mouse was mapped to chromosome 3 by Southern analysis of rodent x mouse hybrid cell DNA. This chromosomal location was confirmed by the use of a restriction fragment length polymorphism (RFLP) in recombinant inbred mouse strains. The results of this RFLP analysis indicated that the mouse Lef-1 gene was closely linked to Pmv-39 and Egf and was likely placed between these loci, both of which were previously mapped to distal mouse chromosome 3. Our mapping results did not suggest involvement of this gene in previously mapped genetic disorders or in known neoplasia-associated translocation breakpoints.

    View details for Web of Science ID A1991GR01600030

    View details for PubMedID 1783375

  • CHARACTERIZATION AND CHROMOSOMAL MAPPING OF A HUMAN STEROID 5-ALPHA-REDUCTASE GENE AND PSEUDOGENE AND MAPPING OF THE MOUSE HOMOLOG GENOMICS JENKINS, E. P., Hsieh, C. L., MILATOVICH, A., Normington, K., Berman, D. M., FRANCKE, U., Russell, D. W. 1991; 11 (4): 1102-1112

    Abstract

    The enzyme steroid 5 alpha-reductase catalyzes the conversion of testosterone into the more powerful androgen, dihydrotestosterone. We previously described the cloning of rat and human cDNAs that encode steroid 5 alpha-reductase and their expression in oocytes and cultured cells. Here, we report the isolation, characterization, and chromosomal mapping of two human steroid 5 alpha-reductase genes. One gene (symbol SRD5A1) is functional, contains five exons separated by four introns, and maps to the distal short arm of chromosome 5. Two informative restriction fragment length polymorphisms are present in exons 1 and 2 of this gene. A second gene (symbol SRD5AP1) has all of the hallmarks of a processed pseudogene and was mapped to the q24-qter region of the X chromosome. In the mouse, a single steroid 5 alpha-reductase gene (Srd5 alpha-1) is linked to Xmv-13 on chromosome 13.

    View details for Web of Science ID A1991GR01600038

    View details for PubMedID 1686016

  • A MELANOCYTE-SPECIFIC GENE, PMEL-17, MAPS NEAR THE SILVER COAT COLOR LOCUS ON MOUSE CHROMOSOME-10 AND IS IN A SYNTENIC REGION ON HUMAN CHROMOSOME-12 PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Kwon, B. S., CHINTAMANENI, C., Kozak, C. A., Copeland, N. G., Gilbert, D. J., Jenkins, N., Barton, D., FRANCKE, U., Kobayashi, Y., Kim, K. K. 1991; 88 (20): 9228-9232

    Abstract

    Melanocytes preferentially express an mRNA species, Pmel 17, whose protein product cross-reacts with anti-tyrosinase antibodies and whose expression correlates with the melanin content. We have now analyzed the deduced protein structure and mapped its chromosomal location in mouse and human. The amino acid sequence deduced from the nucleotide sequence of the Pmel 17 cDNA showed that the protein is composed of 645 amino acids with a molecular weight of 68,600. The Pmel 17 protein contains a putative leader sequence and a potential membrane anchor segment, which indicates that this may be a membrane-associated protein in melanocytes. The deduced protein contains five potential N-glycosylation sites and relatively high levels of serine and threonine. Three repeats of a 26-amino acid motif appear in the middle of the molecule. The human Pmel 17 gene, designated D12S53E, maps to chromosome 12, region 12pter-q21; and the mouse homologue, designated D12S53Eh, maps to the distal region of mouse chromosome 10, a region also known to carry the coat color locus si (silver).

    View details for Web of Science ID A1991GK67600077

    View details for PubMedID 1924386

  • TUMOR-NECROSIS-FACTOR RECEPTOR GENES, TNFR1 AND TNFR2, ON HUMAN CHROMOSOME-12 AND CHROMOSOME-1 SOMATIC CELL AND MOLECULAR GENETICS MILATOVICH, A., Song, K., Heller, R. A., FRANCKE, U. 1991; 17 (5): 519-523

    Abstract

    Tumor necrosis factor, TNF, is a 17-kDa protein secreted by macrophages and classified as a cytokine. TNF binds to high-affinity receptors on the cell surface and is involved in a wide variety of biological responses. There are at least two types of receptors, tumor necrosis factor receptors 1 and 2 (TNFR1 and TNFR2). The genes for TNFR1 a 55-kDa protein, and TNFR2, a 70-kDa protein, have been mapped to human chromosomes 1 12 (12pter-cen) and (1pter-p32), respectively, by Southern blot analysis of human x Chinese hamster somatic cell hybrid panels. Recently, the corresponding genes in the mouse have been mapped to chromosomes 4 and 6 in regions that are conserved on human chromosomes 1 and 12.

    View details for Web of Science ID A1991GZ55100011

    View details for PubMedID 1662415

  • ISOLATION AND CHROMOSOMAL LOCALIZATION OF A NOVEL NONERYTHROID ANKYRIN GENE GENOMICS Tse, W. T., Menninger, J. C., YANGFENG, T. L., FRANCKE, U., Sahr, K. E., Lux, S. E., WARD, D. C., Forget, B. G. 1991; 10 (4): 858-866

    Abstract

    Immunoreactive isoforms of erythrocyte ankyrin have been shown to be present in a variety of nonerythroid tissues. Isolation of the genes that encode these isoforms will clarify their relationship to erythrocyte ankyrin. Using an erythrocyte ankyrin cDNA clone as a hybridization probe, we screened a human genomic library and isolated a clone that hybridizes with the probe at low stringency but not at high stringency. Partial nucleotide sequence of the clone revealed the presence of a 99-bp segment that is homologous to an exon of the erythrocyte ankyrin gene. Northern analysis showed that a labeled fragment of the clone hybridized to a 7-kb message in RNA of fetal brain but not of erythroid cells, suggesting that this clone is part of a novel gene that is expressed predominantly in nonerythroid tissue. Comparison of the sequence of the genomic clone with that of a recently isolated cDNA clone for brain ankyrin (Otto et al., 1989) showed identity of 96 of 99 bp between the putative exon and a segment of the cDNA clone (V. Bennett, personal communication, 1991), suggesting that the genomic clone is part of a gene for nonerythroid ankyrin, which we have designated ANK2. By analysis of somatic cell hybrids and fluorescence in situ hybridization, we assigned ANK2 to human chromosome 4 at a position equivalent to bands 4q25-q27.

    View details for Web of Science ID A1991FW38000002

    View details for PubMedID 1833308

  • GENOMIC ORGANIZATION AND CHROMOSOMAL LOCALIZATION OF THE TAPA-1 GENE JOURNAL OF IMMUNOLOGY Andria, M. L., Hsieh, C. L., Oren, R., FRANCKE, U., Levy, S. 1991; 147 (3): 1030-1036

    Abstract

    TAPA-1 is a 26-kDa integral membrane protein expressed on many human cell types. Antibodies against TAPA-1 induce homotypic aggregation of cells and can inhibit their growth. The murine homologue of TAPA-1 was cloned from both cDNA and genomic DNA libraries. A very high level of homology was found between human and mouse TAPA-1. The 5' untranslated region of the TAPA-1 gene resembles housekeeping gene promoters with respect to G + C content and the presence of potential Sp1 binding sites. The chromosomal localization of human and murine TAPA-1 genes was determined by Southern blot experiments using DNA from somatic cell hybrids. The genes were found to be part of a conserved syntenic group in mouse chromosome 7 and the short arm of human chromosome 11. The organization of the TAPA-1 gene and the projection of the exon boundaries on the proposed protein structure are presented.

    View details for Web of Science ID A1991FY29200041

    View details for PubMedID 1650385

  • CHROMOSOMAL MAPPING OF BRAIN-DERIVED NEUROTROPHIC FACTOR AND NEUROTROPHIN-3 GENES IN MAN AND MOUSE GENOMICS Ozcelik, T., Rosenthal, A., FRANCKE, U. 1991; 10 (3): 569-575

    Abstract

    Brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NTF3) are two new members of the nerve growth factor gene family, which play important roles in the development and maintenance of the vertebrate nervous system. Here we describe the assignments of the BDNF and NTF3 gene loci to human and mouse chromosomes and discuss the evolutionary relationship of human chromosomes 11 and 12. BDNF has been mapped to human chromosome 11p15.5-p11.2 and to mouse chromosome 2, and NTF3 to human chromosome 12p and mouse chromosome 6.

    View details for Web of Science ID A1991FQ64000008

    View details for PubMedID 1889807

  • 242 BREAKPOINTS IN THE 200-KB DELETION-PRONE P20-REGION OF THE DMD GENE ARE WIDELY SPREAD GENOMICS BLONDEN, L. A., Grootscholten, P. M., DENDUNNEN, J. T., Bakker, E., Abbs, S., BOBROW, M., BOEHM, C., Van Broeckhoven, C., Baumbach, L., Chamberlain, J., CASKEY, C. T., Denton, M., Felicetti, L., GALLUZI, G., FISCHBECK, K. H., FRANCKE, U., Darras, B., Gilgenkrantz, H., KAPLAN, J. C., Herrmann, F. H., JUNIEN, C., Boileau, C., LIECHTIGALLATI, S., Lindlof, M., Matsumoto, T., Niikawa, N., Muller, C. R., Poncin, J., Malcolm, S., Robertson, E., Romeo, G., Covone, A. E., Scheffer, H., Schroder, E., Schwartz, M., Verellen, C., Walker, A., WORTON, R., Gillard, E., VANOMMEN, G. J. 1991; 10 (3): 631-639

    Abstract

    Using whole cosmids as probes, we have mapped 242 DMD/BMD deletion breakpoints located in the major deletion hot spot of the DMD gene. Of these, 113 breakpoints were mapped more precisely to individual restriction enzyme fragments in the distal 80 kb of the 170-kb intron 44. An additional 12 breakpoints are distributed over the entire region, with no significant local variation in frequency. Furthermore, deletion sizes vary and are not influenced by the positions of the breakpoints. This argues against a predominant role of one or a few specific sequences in causing frequent rearrangements. It suggests that structural characteristics or a more widespread recombinogenic sequence makes this region so susceptible to deletion. Our study revealed several RFLPs, one of which is a 300-bp insertion/deletion polymorphism. Abnormally migrating junction fragments are found in 81% of the precisely mapped deletions and are highly valuable in the diagnosis of carrier females.

    View details for Web of Science ID A1991FQ64000016

    View details for PubMedID 1679746

  • IDENTITY OF HUMAN LYB-2 AND CD72 AND LOCALIZATION OF THE GENE TO CHROMOSOME-9 EUROPEAN JOURNAL OF IMMUNOLOGY VONHOEGEN, I., Hsieh, C. L., SCHARTING, R., FRANCKE, U., Parnes, J. R. 1991; 21 (6): 1425-1431

    Abstract

    We have recently reported the isolation of cDNA clones encoding the human homolog of the mouse B cell differentiation antigen Lyb-2. Expression of Lyb-2 is restricted to B lineage cells and turned off in antibody-secreting plasma cells in both mice and humans. Functional studies with anti-mouse Lyb-2 monoclonal antibodies (mAb) suggest that this protein may be involved in signals for B cell proliferation. We now describe the generation of mAb specific for human Lyb-2. Furthermore, we demonstrate that the human Lyb-2 protein is recognized by a mAb specific for the newly clustered pan B cell surface antigen CD72, which had been defined by mAb. Mouse L(tk-) cells expressing a transfected cDNA encoding human Lyb-2 bind a mAb specific for CD72. We further show that an antiserum and a mAb specific for human Lyb-2 and an anti-CD72 mAb immunoprecipitate the identical protein from human splenic B cells and B cell lines, as well as from transfected L(tk-) cells. These data indicate that CD72 is the human equivalent of mouse Lyb-2. We have additionally localized the gene for human Lyb-2/CD72 on the short arm of chromosome 9. Mouse Lyb-2 had been previously mapped to mouse chromosome 4. Lyb-2/CD72 is now the tenth gene known to be localized on human chromosome 9 and mouse chromosome 4.

    View details for Web of Science ID A1991FT00500014

    View details for PubMedID 2044654

  • SEQUENCE-ANALYSIS, EXPRESSION AND CHROMOSOMAL LOCALIZATION OF A GENE, ISOLATED FROM A SUBTRACTED HUMAN RETINA CDNA LIBRARY, THAT ENCODES AN INSULIN-LIKE GROWTH-FACTOR BINDING-PROTEIN (IGFBP2) EXPERIMENTAL EYE RESEARCH Agarwal, N., Hsieh, C. L., Sills, D., Swaroop, M., Desai, B., FRANCKE, U., Swaroop, A. 1991; 52 (5): 549-561

    Abstract

    The metabolic functions of insulin-like growth factors (IGFs) I and II are modulated by a family of binding proteins which are present in biological fluids and are synthesized by a variety of cell types. A cDNA clone, isolated at random from a subtracted human retina library, has been identified to code for a novel IGF-binding protein (IGFBP2) by its sequence homology to the peptide sequence of IGF binding proteins purified from bovine MDBK and rat BRL-3A cells. The complete nucleotide sequence of the IGFBP2 cDNA is 1406 bp long, contains 66% G-Cs and an open reading frame of 328 amino acids with a putative signal or pro-peptide of 39 residues. The mature polypeptide of 289 amino acids has 18 cysteines, a putative ATP-binding site and an RGD tripeptide. The 1.4 kb IGFBP2 transcript is expressed in several human tissues including fetal eye and fetal brain, but not in the human lymphoblastoid cell line against which the retinal cDNA library was subtracted. In situ hybridization to sections of mouse retina localized the mRNA for IGFBP2 primarily in the outer nuclear layer of photoreceptors. Southern blot analysis of DNA from human x rodent and mouse x rodent somatic cell hybrids assigned the gene for IGFBP2 to human chromosome 2q33-qter and mouse chromosome 1 in a known conserved syntenic region.

    View details for Web of Science ID A1991FP33100008

    View details for PubMedID 1712312

  • STRUCTURE OF THE HUMAN CYTOCHROME-C-OXIDASE SUBUNIT-VB GENE AND CHROMOSOMAL MAPPING OF THE CODING GENE AND OF 7 PSEUDOGENES GENOMICS Lomax, M. I., Hsieh, C. L., DARRAS, B. T., FRANCKE, U. 1991; 10 (1): 1-9

    Abstract

    Subunit Vb of mammalian cytochrome c oxidase (COX; EC 1.9.3.1) is encoded by a nuclear gene and assembled with the other 12 COX subunits encoded in both mitochondrial and nuclear DNA. We have cloned the gene for human COX subunit Vb (COX5B) and determined the exon-intron structure by both hybridization analysis and DNA sequencing. The gene contains five exons and four introns; the four coding exons span a region of approximately 2.4 kb. The 5' end of the COX5B gene is GC-rich and contains many HpaII sites. Genomic Southern blot analysis of human DNA probed with the human COX Vb cDNA identified eight restriction fragments containing COX Vb-related sequences that were mapped to different chromosomes with panels of human x Chinese hamster somatic cell hybrids. Because only one of these fragments hybridized with a 210-bp probe from intron 4, we conclude that there is a single expressed gene for COX subunit Vb in the human genome. We have mapped this gene to chromosome 2, region cen-q13.

    View details for Web of Science ID A1991FK40800001

    View details for PubMedID 1646156

  • MUTATIONS IN THE BILE-ACID BIOSYNTHETIC ENZYME STEROL 27-HYDROXYLASE UNDERLIE CEREBROTENDINOUS XANTHOMATOSIS JOURNAL OF BIOLOGICAL CHEMISTRY Cali, J. J., Hsieh, C. L., FRANCKE, U., Russell, D. W. 1991; 266 (12): 7779-7783

    Abstract

    The sterol storage disorder cerebrotendinous xanthomatosis (CTX) is characterized by abnormal deposition of cholesterol and cholestanol in multiple tissues. Deposition in the central nervous system leads to neurological dysfunction marked by dementia, spinal cord paresis, and cerebellar ataxia. Deposition in other tissues causes tendon xanthomas, premature atherosclerosis, and cataracts. In two unrelated patients with CTX, we have identified different point mutations in the gene (CYP27) encoding sterol 27-hydroxylase, a key enzyme in the bile acid biosynthesis pathway. Transfection of mutant cDNAs into cultured cells results in the synthesis of immunoreactive sterol 27-hydroxylase protein with greatly diminished enzyme activity. We have localized the CYP27 gene to the q33-qter interval of human chromosome 2, and to mouse chromosome 1, in agreement with the autosomal recessive inheritance pattern of CTX. These findings underscore the essential role played by sterols in the central nervous system and suggest that mutations in other sterol metabolizing enzymes may contribute to diseases with neurological manifestations.

    View details for Web of Science ID A1991FJ34200074

    View details for PubMedID 2019602

  • DNA-SEQUENCE, GENOMIC ORGANIZATION, AND CHROMOSOMAL LOCALIZATION OF THE MOUSE PERIPHERAL MYELIN PROTEIN ZERO GENE - IDENTIFICATION OF POLYMORPHIC ALLELES GENOMICS You, K. H., Hsieh, C. L., Hayes, C., Stahl, N., FRANCKE, U., Popko, B. 1991; 9 (4): 751-757

    Abstract

    We have cloned and characterized the mouse gene, P0, that encodes the predominant protein of peripheral myelin. Similar to the rat gene, the mouse P0 gene is encoded by six exons that span about 7 kb of DNA. The DNA sequence of the mouse gene is highly homologous with the rat gene, including the regions believed to be important in transcriptional control. Furthermore, the P0 protein appears well conserved throughout evolution. The gene was mapped to mouse chromosome 1 by Southern analysis of a Chinese hamster x mouse somatic cell hybrid panel. Several polymorphic restriction enzyme sites were identified within the P0 locus. Recombinant inbred strain mapping has linked the P0 gene to Ly-9/Ly/Sap in a region corresponding to band 1H3.

    View details for Web of Science ID A1991FB62900024

    View details for PubMedID 1709914

  • CDNA AND GENOMIC CLONING OF HRC, A HUMAN SARCOPLASMIC-RETICULUM PROTEIN, AND LOCALIZATION OF THE GENE TO HUMAN CHROMOSOME-19 AND MOUSE CHROMOSOME-7 GENOMICS Hofmann, S. L., Topham, M., Hsieh, C. L., FRANCKE, U. 1991; 9 (4): 656-669

    Abstract

    Histidine-rich calcium binding protein (HRC) is a luminal sarcoplasmic reticulum (SR) protein of 165 kDa identified by virtue of its ability to bind 125I-labeled low-density lipoprotein with high affinity after sodium dodecyl sulfate-polyacrylamide gel electrophoresis (Hofmann et al., J. Biol. Chem. 264: 8260-8270, 1989). Its role in SR function is unknown. In this report, the gene encoding human HRC was localized to human chromosome 19 and mouse chromosome 7 by hybridization of a human HRC cDNA fragment to a panel of somatic cell hybrids. Known synteny between a portion of human chromosome 19 and a portion of mouse chromosome 7 and in situ hybridization of a biotin-labeled HRC probe to human chromosomes suggest a localization to a region corresponding to 19q13.3. The locus for myotonic dystrophy resides in the region 19q13.2-13.3. Therefore, we considered HRC, a muscle-specific gene, to possibly represent a "candidate gene" for myotonic muscular dystrophy. As a first step toward localizing HRC in relation to the myotonic dystrophy locus, we report the cloning of the human HRC gene, its intron-exon organization, and characterization of several informative polymorphisms to be used in future linkage studies in families with myotonic dystrophy. Of particular interest is an Alu-associated poly-d(GA) sequence located in an intron in the middle of the gene, and two stretches of acidic amino acids in the coding region of exon 1 that vary in length among different individuals.

    View details for Web of Science ID A1991FB62900013

    View details for PubMedID 2037293

  • RETT SYNDROME - EXCLUSION MAPPING FOLLOWING THE HYPOTHESIS OF GERMINAL MOSAICISM FOR NEW X-LINKED MUTATIONS HUMAN GENETICS Archidiacono, N., Lerone, M., Rocchi, M., ANVRET, M., Ozcelik, T., FRANCKE, U., Romeo, G. 1991; 86 (6): 604-606

    Abstract

    The hypothesis of germinal mosaicism in the unaffected mother of two half-sisters affected with Rett syndrome is postulated to explain the unusual recurrence of this genetic disorder affecting only females (1/10,000); it might be caused by new X-linked mutations with lethality in male fetuses. The analysis of 34 X-linked restriction fragment length polymorphisms (RFLPs) in these two affected females and in their unaffected mother and half-brother, together with the reconstruction of phase for 15 informative RFLPs in somatic cell hybrids retaining a single X chromosome from each female, has made it possible to exclude some regions of the X chromosome as possible sites of the mutation(s) causing Rett syndrome.

    View details for Web of Science ID A1991FH81000012

    View details for PubMedID 1673961

  • GUIDELINES FOR HUMAN LINKAGE MAPS - AN INTERNATIONAL SYSTEM FOR HUMAN LINKAGE MAPS (ISLM, 1990) GENOMICS Keats, B. J., Sherman, S. L., Morton, N. E., ROBSON, E. B., Buetow, K. H., CARTWRIGHT, P. E., Chakravarti, A., FRANCKE, U., GREEN, P. P., Ott, J. 1991; 9 (3): 557-560

    View details for Web of Science ID A1991EY81600021

    View details for PubMedID 2032725

  • DISTRIBUTION OF GENES FOR GAP JUNCTION MEMBRANE CHANNEL PROTEINS ON HUMAN AND MOUSE CHROMOSOMES SOMATIC CELL AND MOLECULAR GENETICS Hsieh, C. L., KUMAR, N. M., GILULA, N. B., FRANCKE, U. 1991; 17 (2): 191-200

    Abstract

    Gap junctions are widely distributed structures that mediate communication between cells. The channels that allow passage of small molecules between adjacent cells are made up of oligomeric proteins (connexins) that are encoded by a family of related genes. By probing somatic cell hybrid DNA on Southern filters with rat or human cDNAs or human genomic fragments, we have mapped four functioning gap junction genes, (alpha 1, beta 1, beta 2, and alpha 3), to different sites on human chromosomes: GJA1 (connexin43) to 6p21.1-q24.1; GJB1 (connexin32) to Xcen-q22; GJB2 (connexin26) to 13; and GJA3 (connexin46) also to 13, probably near GJB2. The GJA3 probe also hybridized to a restriction fragment that was mapped to chromosome 1. A GJA1-related pseudogene GJA1P was assigned to chromosome 5. The homologous loci in mouse were assigned to regions of known conserved syntenic groups: Gja-1 to chromosome 10; Gjb-1 to XD-F4 and Gjb-2 to 14. Of two sites of hybridization with the GJA3 probe, on mouse 14 and 5, we assume that the site on 14 corresponds to the GJA3 locus on human 13. Based on these data, additional members of this family of related genes can be isolated and characterized, and possible human and mouse mutations can be identified.

    View details for Web of Science ID A1991FD59400008

    View details for PubMedID 1849321

  • HOMOZYGOUS TYROSINASE GENE MUTATION IN AN AMERICAN BLACK WITH TYROSINASE-NEGATIVE (TYPE-IA) OCULOCUTANEOUS ALBINISM AMERICAN JOURNAL OF HUMAN GENETICS Spritz, R. A., STRUNK, K. M., Hsieh, C. L., Sekhon, G. S., FRANCKE, U. 1991; 48 (2): 318-324

    Abstract

    We have identified a tyrosinase gene mutation in an American black with classic, tyrosinase-negative oculocutaneous albinism. This mutation results in an amino acid substitution (Cys----Arg) at codon 89 of the tyrosinase polypeptide. The proband is homozygous for the substitution, suggesting that this mutation may be frequently associated with tyrosinase-negative oculocutaneous albinism in blacks.

    View details for Web of Science ID A1991EV74400016

    View details for PubMedID 1899321

  • PROPERTIES OF TITIN Labeit, S., BARLOW, D. P., Gautel, M., Gibson, T., Holt, J., Hsieh, C. L., FRANCKE, U., Leonard, K., Gamage, M., Phillips, A., WARDALE, J., Whiting, A., Trinick, J. SPRINGER. 1991: 115–15
  • STRUCTURAL AND FUNCTIONAL CONSERVATION OF SYNAPTOTAGMIN (P65) IN DROSOPHILA AND HUMANS JOURNAL OF BIOLOGICAL CHEMISTRY Perin, M. S., Johnston, P. A., Ozcelik, T., Jahn, R., FRANCKE, U., Sudhof, T. C. 1991; 266 (1): 615-622

    Abstract

    Synaptotagmin (p65) is an abundant synaptic vesicle protein that contains two copies of a sequence that is homologous to the regulatory region of protein kinase C. Full length cDNAs encoding human and Drosophila synaptotagmins were characterized to study its structural and functional conservation in evolution. The deduced amino acid sequences for human and rat synaptotagmins show 97% identity, whereas Drosophila and rat synaptotagmins are only 57% identical but exhibit a selective conservation of the two internal repeats that are homologous to the regulatory region of protein kinase C (78% invariant residues in all three species). The two internal repeats of synaptotagmin are only slightly more homologous to each other than to protein kinase C, and the differences between the repeats are conserved in evolution, suggesting that they might not be functionally equivalent. The cytoplasmic domains of human and Drosophila synaptotagmins produced as recombinant proteins in Escherichia coli specifically bound phosphatidylserine similar to rat synaptotagmin. They also hemagglutinated trypsinized erythrocytes at nanomolar concentrations. Hemagglutination was inhibited both by negatively charged phospholipids and by a recombinant fragment from rat synaptotagmin that contained only a single copy of the two internal repeats. Together these results demonstrate that synaptotagmin is highly conserved in evolution compatible with a function in the trafficking of synaptic vesicles at the active zone. The similarity of the phospholipid binding properties of the cytoplasmic domains of rat, human, and Drosophila synaptotagmins and the selective conservation of the sequences that are homologous to protein kinase C suggest that these are instrumental in phospholipid binding. The human gene for synaptotagmin was mapped by Southern blot analysis of DNA from somatic cell hybrids to chromosome 12 region cen-q21, and the Drosophila gene by in situ hybridization to 23B.

    View details for Web of Science ID A1991EQ33900089

    View details for PubMedID 1840599

  • CHROMOSOMAL LOCALIZATION OF THE GENE FOR AA-TYPE PLATELET-DERIVED GROWTH-FACTOR RECEPTOR (PDGFRA) IN HUMANS AND MICE CYTOGENETICS AND CELL GENETICS Hsieh, C. L., NAVANKASATTUSAS, S., Escobedo, J. A., Williams, L. T., FRANCKE, U. 1991; 56 (3-4): 160-163

    Abstract

    The full-length cDNA of the receptor for human AA-type platelet-derived growth factor (PDGF) was used to assign the PDGFRA gene to region q11----q21 of human chromosome 4 and to mouse Chromosome 5 by somatic cell hybrid analysis. Since the same region also contains the c-kit oncogene homolog KIT, we carried out pulsed-field gel electrophoresis to determine the physical distance between the two genes in human DNA. The two probes, when successively applied to the same filters, hybridized to a 450-kb EagI-fragment but not to other common restriction fragments. The genes are separated by at least one NotI, one XhoI, and one SalI site.

    View details for PubMedID 1711435

  • THE GENES FOR 2 TUMOR-NECROSIS-FACTOR RECEPTORS, TNFR1 AND TNFR2, MAP TO HUMAN CHROMOSOME-12 (12PTER-CEN) AND CHROMOSOME-1 (1PTER-P-32) MILATOVICH, A., Song, K., Heller, R. A., FRANCKE, U. KARGER. 1991: 1980–80
  • ASSIGNMENT OF THE HUMAN SYNDECAN (SDC) GENE TO SHORT ARM OF CHROMOSOME-2 Westman, P., Hsieh, C. L., Mali, M., JALKANEN, M., FRANCKE, U., Schroder, J. KARGER. 1991: 1873–74
  • CHROMOSOMAL MAPPING OF HUMAN STEROID-5 ALPHA-REDUCTASE GENE (SRD5A1) AND PSEUDOGENE (SRD5AP1) IN HUMAN AND MOUSE Hsieh, C. L., MILATOVICH, A., Russell, D., FRANCKE, U. KARGER. 1991: 1897–97
  • SEQUENCES HOMOLOGOUS TO MTTF1, MITOCHONDRIAL TRANSCRIPTION FACTOR-I, ARE LOCATED ON HUMAN CHROMOSOME-7 (7PTER-CEN), CHROMOSOME-10 AND CHROMOSOME-11 (11CEN-QTER) MILATOVICH, A., Parisi, M. A., Poulton, J., Clayton, D. A., FRANCKE, U. KARGER. 1991: 1924–24
  • GUIDELINES FOR HUMAN LINKAGE MAPS - AN INTERNATIONAL SYSTEM FOR HUMAN LINKAGE MAPS (ISLM, 1990) ANNALS OF HUMAN GENETICS Keats, B. J., Sherman, S. L., Morton, N. E., ROBSON, E. B., Buetow, K. H., CARTWRIGHT, P. E., Chakravarti, A., FRANCKE, U., GREEN, P. P., Ott, J. 1991; 55: 1-6

    View details for Web of Science ID A1991FD02000001

    View details for PubMedID 2042931

  • Cytogenetic analysis in melanoma and nevi. Cancer treatment and research Cowan, J. M., FRANCKE, U. 1991; 54: 3-16

    Abstract

    We have presented data from the cytogenetic analysis of melanocytes derived from four dysplastic nevi and 14 melanomas. We have confirmed previous results from melanomas, including the loss of chromosome 6q and duplication of 1q and 7p. We did not observe an excess of rearrangements involving chromosomes 2 and 3, but we found frequent deletion of chromosomes 9p, 10p, 10q, and 11q. We observed the loss of chromosome 9 in 2 of 4 dysplastic nevi, and combining this observation with that from the melanomas suggests that deletion or inactivation of a gene on 9p may be a primary event in melanocyte transformation. Other tumor suppressor genes are likely to be involved in the transformation process, and these are most likely located on 10p, 10q, 11q, and 6q. We observed many rearrangement involving chromosome band 1p13 and suggest that activation of a gene in that band may also contribute to the transformation process.

    View details for PubMedID 1673858

  • MOLECULAR-CLONING, FUNCTIONAL EXPRESSION, AND CHROMOSOMAL LOCALIZATION OF MOUSE HEPATOCYTE NUCLEAR FACTOR-I PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Kuo, C. J., Conley, P. B., Hsieh, C. L., FRANCKE, U., Crabtree, G. R. 1990; 87 (24): 9838-9842

    Abstract

    The homeodomain-containing transcription factor hepatocyte nuclear factor 1 (HNF-1) most likely plays an essential role during liver organogenesis by transactivating a family of greater than 15 predominantly hepatic genes. We have isolated cDNA clones encoding mouse HNF-1 and expressed them in monkey COS cells and in the human T-cell line Jurkat, producing HNF-1 DNA-binding activity as well as transactivation of reporter constructs containing multimerized HNF-1 binding sites. In addition, the HNF-1 gene was assigned by somatic cell hybrids and recombinant inbred strain mapping to mouse chromosome 5 near Bcd-1 and to human chromosome 12 region q22-qter, revealing a homologous chromosome region in these two species. The presence of HNF-1 mRNA in multiple endodermal tissues (liver, stomach, intestine) suggests that HNF-1 may constitute an early marker for endodermal, rather than hepatocyte, differentiation. Further, that HNF-1 DNA-binding and transcriptional activity can be conferred by transfecting the HNF-1 cDNA into several cell lines indicates that it is sufficient to activate transcription in the context of ubiquitously expressed factors.

    View details for Web of Science ID A1990EN15900069

    View details for PubMedID 2263635

  • THE SEROTONIN RECEPTOR SUBTYPE-2 LOCUS HTR2 IS ON HUMAN CHROMOSOME-13 NEAR GENES FOR ESTERASE-D AND RETINOBLASTOMA-1 AND ON MOUSE CHROMOSOME-14 SOMATIC CELL AND MOLECULAR GENETICS Hsieh, C. L., Bowcock, A. M., Farrer, L. A., Hebert, J. M., Huang, K. N., CAVALLISFORZA, L. L., Julius, D., FRANCKE, U. 1990; 16 (6): 567-574

    Abstract

    Serotonin (5-hydroxytryptamine) functions as a neurotransmitter and a hormone. Its diverse actions are mediated by at least seven distinct cell surface receptor subtypes. The serotonin receptor subtype 2 (gene symbol HTR2) is a G-protein-coupled receptor, expressed primarily in the cerebral cortex, where upon stimulation it stimulates the hydrolysis of inositol phospholipids. We have mapped the HTR2 locus to human chromosome 13 and to mouse chromosome 14 by somatic cell hybrid analysis. Linkage studies in CEPH families, using a PvuII RFLP detected with the HTR2 probe, revealed tight linkage between HTR2 and ESD, the locus for esterase D. The most likely position for HTR2 is between ESD and RB1, the retinoblastoma-1 gene. The homologous loci in mouse, Rb-1 and Esd(Es-10) are on mouse chromosome 14, close to ag, agitans, a recessive neurological mutation. Having mapped Htr-2 to mouse chromosome 14, we predict that it falls into this known conserved gene cluster.

    View details for Web of Science ID A1990ER23200007

    View details for PubMedID 1980030

  • STRUCTURES AND CHROMOSOMAL LOCALIZATIONS OF 2 HUMAN GENES ENCODING SYNAPTOBREVIN-1 AND SYNAPTOBREVIN-2 JOURNAL OF BIOLOGICAL CHEMISTRY ARCHER, B. T., Ozcelik, T., Jahn, R., FRANCKE, U., Sudhof, T. C. 1990; 265 (28): 17267-17273

    Abstract

    Synaptobrevins 1 and 2 are small integral membrane proteins specific for synaptic vesicles in neurons. Two cosmid clones containing the human genes encoding synaptobrevins 1 and 2 (gene symbols SYB1 and SYB2, respectively) were isolated and characterized. The coding regions of the synaptobrevin genes are highly homologous to each other and are interrupted at identical positions by introns of different size and sequence. Each gene is organized into five exons whose boundaries correspond to those of the protein domains. Exon I contains part of the initiator methionine codon whereas exon II encodes the variable and immunogenic amino-terminal domain of the synaptobrevins. The third exon comprises the highly conserved central domain of the synaptobrevins, exon IV encodes most of the transmembrane region, and exon V contains the last residues of the transmembrane region and the small intravesicular carboxyl terminus. Comparisons of the synaptobrevin sequences in five species from Drosophila with man indicate a selective conservation of sequences adjacent to the synaptic vesicle surface, suggesting a function at the membrane-cystosol interface. The chromosomal localizations of the human and mouse SYB1 and SYB2 genes were determined using hybrid cell lines. SYB1 was localized to the short arm of human chromosome 12 and to mouse chromosome 6 whereas SYB2 was found on the distal portion of the short arm of human chromosome 17 and on mouse chromosome 11. A PstI restriction fragment length polymorphism was identified at the SYB2 locus.

    View details for Web of Science ID A1990EA85700086

    View details for PubMedID 1976629

  • MOLECULAR-STRUCTURE, CHROMOSOME ASSIGNMENT, AND PROMOTER ORGANIZATION OF THE HUMAN MATRIX GLA PROTEIN GENE JOURNAL OF BIOLOGICAL CHEMISTRY Cancela, L., Hsieh, C. L., FRANCKE, U., Price, P. A. 1990; 265 (25): 15040-15048

    Abstract

    Matrix Gla protein (MGP) is an 84-residue vitamin K-dependent protein initially isolated from bovine bone. MGP is also expressed at high levels in heart, kidney, and lung and is up-regulated by vitamin D in bone cells. To characterize the genomic sequences responsible for the regulated expression of this gene, we screened a human genomic library using a MGP cDNA probe and obtained two clones containing the MGP locus. The human MGP gene spans 3.9 kilobases of chromosomal DNA and consists of four exons separated by three large intervening sequences which account for more than 80% of the gene. Southern analysis of total human genomic DNA indicated the presence of a single copy of the MGP gene. Hybridization of the hMGP cDNA to a series of Chinese Hamster x human hybrid clones assigned this gene to the short arm of the human chromosome 12 (12p). The N-terminal sequences of the known vitamin K-dependent vertebrate proteins reveal a transmembrane signal peptide, followed by a putative gamma-carboxylation recognition site and a Gla-containing domain. Each of these regions correspond to a separate exon in MGP. MGP also contains a fourth exon of unknown function which codes for 11 residues and lies between the transmembrane signal peptide and the putative recognition site for the gamma-carboxylase. This four-exon organization is essentially identical to that of bone Gla protein and is quite different from the two exon organization encoding this region in the other known vitamin K-dependent proteins. Analysis of the MGP gene promoter revealed, in addition to the typical TATA and CAT boxes, the presence of a number of putative regulatory sequences homologous to previously identified hormone and transcription factor responsive elements. In particular, two regions of the promoter were delineated containing possible binding sites for retinoic acid and vitamin D receptors.

    View details for Web of Science ID A1990DX40000059

    View details for PubMedID 2394711

  • GENES FOR 2 AUTOSOMAL RECESSIVE FORMS OF CHRONIC GRANULOMATOUS-DISEASE ASSIGNED TO 1Q25 (NCF2) AND 7Q11.23 (NCF1) AMERICAN JOURNAL OF HUMAN GENETICS FRANCKE, U., Hsieh, C. L., FOELLMER, B. E., LOMAX, K. J., Malech, H. L., Leto, T. L. 1990; 47 (3): 483-492

    Abstract

    Chronic granulomatous disease (CGD) is a heterogeneous group of inherited disorders of impaired superoxide production in phagocytes. The most common X-linked recessive form involves the CYBB locus in band Xp21.1 that encodes the membrane-bound beta subunit of the cytochrome b558 complex. Two autosomal recessive forms of CGD result from defects in cytosolic components of the phagocyte NADPH oxidase system, p47phox (NCF1) and p67phox (NCF2). By using human cDNA probes we have mapped the genes for these proteins to chromosomal sites. The combined data from Southern analysis of somatic cell hybrid lines and chromosomal in situ hybridization localize NCF1 to 7q11.23 and NCF2 to band 1q25. The NCF1 localization corrects an erroneous preliminary assignment to chromosome 10. In the mouse, the locus corresponding to NCF2 (Ncf-2) was mapped with somatic cell hybrid panels and recombinant inbred strains to mouse chromosome 1 near Xmv-21 within a region of conserved homology with human chromosome 1 region q21-q32. A second site, probably a processed pseudogene, was identified on mouse chromosome 13.

    View details for Web of Science ID A1990DX22300013

    View details for PubMedID 2393022

  • SYNAPTOPHYSIN - STRUCTURE OF THE HUMAN GENE AND ASSIGNMENT TO THE X-CHROMOSOME IN MAN AND MOUSE AMERICAN JOURNAL OF HUMAN GENETICS Ozcelik, T., Lafreniere, R. G., ARCHER, B. T., Johnston, P. A., WILLARD, H. F., FRANCKE, U., Sudhof, T. C. 1990; 47 (3): 551-561

    Abstract

    Synaptophysin is an integral membrane protein of small synaptic vesicles in brain and endocrine cells. We have determined the structure and organization of the human synaptophysin gene and have established the chromosome localizations in man and mouse. Analysis of a cosmid clone containing the human synaptophysin gene (SYP) revealed seven exons distributed over approximately 20 kb, when compared with the previously published cDNA sequence. The exon-intron boundaries have been identified and do not correlate with functional domains. One intron interrupts the 3' untranslated region. Chromosomal localization of the human and murine genes for synaptophysin established the human SYP locus on the X chromosome in subbands Xp11.22-p11.23 and the mouse synaptophysin gene locus (Syp) on the X chromosome in region A-D. In addition, an Eco0109 RFLP has been identified and used in genetic mapping of the human SYP locus and supports the order TIMP-SYP-DXS14 within a span of approximately 4-7 centimorgans.

    View details for Web of Science ID A1990DX22300020

    View details for PubMedID 1975480

  • ORIGIN OF MUTATIONS IN 2 FAMILIES WITH X-LINKED CHRONIC GRANULOMATOUS-DISEASE BLOOD FRANCKE, U., Ochs, H. D., DARRAS, B. T., Swaroop, A. 1990; 76 (3): 602-606

    Abstract

    The most common X-linked recessive form of chronic granulomatous disease (X-CGD) is characterized by the absence of cytochrome b558 in neutrophils. In a rare variant form of X-CGD, cytochrome b558 is present but not functional. The gene (locus symbol CYBB) was localized to band Xp21 by studies of patients with small chromosome deletions. The gene was cloned based on its location and found to encode the 91-Kd subunit of the cytochrome b558 complex. Most female carriers for X-CGD can be identified by their X-inactivation mosaicism; on average 50% of their neutrophils express the mutant phenotype and fail to reduce nitroblue tetrazolium (NBT). In 2 of 4 families studied, the maternal grandmothers had normal NBT tests, suggesting either nonrandom X-inactivation or new mutations. Restriction fragment length polymorphism analysis using closely linked flanking markers or the NsiI polymorphism detected by the CYBB probe itself, allowed us to identify the X chromosome carrying the mutation as derived from a healthy NBT-positive maternal grandfather. The mothers of the affected boys must have received a paternal X chromosome carrying a new mutation, consistent with the maternal grandmothers' normal NBT tests. In all of eight potential carriers studied, the results of the NBT and DNA marker testing were in complete agreement. Prenatal diagnosis by DNA testing can be performed in early gestation obviating the need for fetal blood sampling.

    View details for Web of Science ID A1990DR56700024

    View details for PubMedID 1974155

  • INSULIN-DEGRADING ENZYME - STABLE EXPRESSION OF THE HUMAN COMPLEMENTARY-DNA, CHARACTERIZATION OF ITS PROTEIN PRODUCT, AND CHROMOSOMAL MAPPING OF THE HUMAN AND MOUSE GENES MOLECULAR ENDOCRINOLOGY Affholter, J. A., Hsieh, C. L., FRANCKE, U., Roth, R. A. 1990; 4 (8): 1125-1135

    Abstract

    We have recently described the isolation of a cDNA encoding an enzyme thought to be involved in the degradation of insulin by insulin-responsive tissues. This enzyme, referred to as insulin-degrading enzyme (IDE), is a cytosolic proteinase of 110,000 mol wt which shares structural and functional homology with bacterial protease III. The enzyme may function in the termination of the insulin response. We report here the mapping of the human and mouse IDE genes to human chromosome 10 and mouse chromosome 19, respectively, and evidence for the existence of a single complex IDE gene. We also describe the stable transfection of Chinese hamster ovary cells with a plasmid containing the IDE cDNA under the transcriptional control of the SR alpha promoter. The recombinant protein synthesized by these cells is indistinguishable from the isolated human enzyme in both its size and immunoreactivity and degrades insulin with a specific activity similar to that of the purified proteinase. Overexpression of IDE using this system should allow for a functional test of the role of IDE in insulin action. In addition, expression of various site-directed mutants of IDE will aid in identifying the residues of IDE and protease III that are essential to the activity of this unique family of proteinases.

    View details for Web of Science ID A1990DU70300005

    View details for PubMedID 2293021

  • CHROMOSOMAL LOCALIZATION AND CDNA SEQUENCE OF HUMAN RALB, A GTP BINDING-PROTEIN SOMATIC CELL AND MOLECULAR GENETICS Hsieh, C. L., Swaroop, A., FRANCKE, U. 1990; 16 (4): 407-410

    Abstract

    The human ra1 proteins share more than 50% homology and some properties with the ras proteins. The RALB gene, a new member of the ra1 family, has been isolated, sequenced, and assigned to human chromosome 2 cen-q13. One other member of this family, RALA, has been previously mapped to the short arm of human chromosome 7.

    View details for Web of Science ID A1990EB28000013

    View details for PubMedID 2120779

  • ASSIGNMENT OF AMOG (ADHESION MOLECULE ON GLIA) GENE TO MOUSE CHROMOSOME-11 NEAR ZFP-3 AND ASGR-1,2 AND TO HUMAN CHROMOSOME-17 SOMATIC CELL AND MOLECULAR GENETICS Hsieh, C. L., CHENGDEUTSCH, A., Gloor, S., Schachner, M., FRANCKE, U. 1990; 16 (4): 401-405

    Abstract

    AMOG, identified as an adhesion molecule that mediates neuron-astrocyte interaction, has structural similarity to the beta-subunit of Na,K ATPase. We have mapped the AMOG gene to human chromosome 17 and mouse chromosome 11 by somatic cell hybrid analysis. Recombinant inbred strain mapping has placed the Amog locus close to genes for zinc finger protein-3 and the asialoglycoprotein receptor in a region of mouse chromosome 11 that is homologous to human 17p.

    View details for Web of Science ID A1990EB28000012

    View details for PubMedID 1699290

  • CD14 IS A MEMBER OF THE FAMILY OF LEUCINE-RICH PROTEINS AND IS ENCODED BY A GENE SYNTENIC WITH MULTIPLE RECEPTOR GENES JOURNAL OF IMMUNOLOGY Ferrero, E., Hsieh, C. L., FRANCKE, U., Goyert, S. M. 1990; 145 (1): 331-336

    Abstract

    We have isolated and characterized genomic and cDNA clones encoding the murine homolog of the human monocyte/granulocyte cell surface glycoprotein, CD14. As in man, the expression of murine CD14 is limited to the myeloid lineage. The murine and human CD14 genes are highly conserved in their intron-exon organization and nucleotide sequence. Their deduced protein sequences show 66% amino acid identity. In both mouse and man, the CD14 protein contains a repeating (10 times) leucine-rich motif (LXXLXLX) that is also found in a group of heterogeneous proteins from phylogenetically distant species. The CD14 gene has been mapped to mouse chromosome 18 which also contains at least five genes encoding receptors (Pdgfr, Adrb2r, li, Grl-1, Fms). Thus CD14 and the receptor genes form a conserved syntenic group localized on mouse chromosome 18 and human chromosome 5. The inclusion of CD14 in the family of leucine-rich proteins, its expression profile and the murine chromosomal localization support the hypothesis that CD14 may function as a receptor.

    View details for Web of Science ID A1990DL17800046

    View details for PubMedID 1694207

  • HEREDITARY SPHEROCYTOSIS ASSOCIATED WITH DELETION OF HUMAN ERYTHROCYTE ANKYRIN GENE ON CHROMOSOME-8 NATURE Lux, S. E., Tse, W. T., Menninger, J. C., John, K. M., Harris, P., Shalev, O., Chilcote, R. R., Marchesi, S. L., WATKINS, P. C., Bennett, V., McIntosh, S., Collins, F. S., FRANCKE, U., WARD, D. C., Forget, B. G. 1990; 345 (6277): 736-739

    Abstract

    Hereditary spherocytosis (HS) is one of the most common hereditary haemolytic anaemias. HS red cells from both autosound dominant and recessive variants are spectrin-deficient, which correlates with the severity of the disease. Some patients with recessive HS have a mutation in the spectrin alpha-2 domain (S.L.M. et al., unpublished observations), and a few dominant HS patients have an unstable beta-spectrin that is easily oxidized, which damages the protein 4.1 binding site and weakens spectrin-actin interactions. In most patients, however, the cause of spectrin deficiency is unknown. The alpha- and beta-spectrin loci are on chromosomes 1 and 14 respectively. The only other genetic locus for HS is SPH2, on the short arm of chromosome 8 (8p11). This does not correspond to any of the known loci of genes for red cell membrane proteins including protein 4.1 (1p36.2-p34), the anion exchange protein (AE1, band 3; 17q21-qter), glycophorin C (2q14-q21), and beta-actin (7pter-q22). Human erythrocyte ankyrin, which links beta-spectrin to the anion exchange protein, has recently been cloned. We now show that the ankyrin gene maps to chromosome 8p11.2, and that one copy is missing from DNA of two unrelated children with severe HS and heterozygous deletions of chromosome 8 (del(8)(p11-p21.1)). Affected red cells are also ankyrin-deficient. The data suggest that defects or deficiency or ankyrin are responsible for HS at the SPH2 locus.

    View details for Web of Science ID A1990DK39900074

    View details for PubMedID 2141669

  • HUMAN EMBRYONIC ATRIAL MYOSIN ALKALI LIGHT CHAIN GENE - CHARACTERIZATION, SEQUENCE, AND CHROMOSOMAL LOCATION GENOMICS Seharaseyon, J., Bober, E., Hsieh, C. L., Fodor, W. L., FRANCKE, U., Arnold, H. H., Vanin, E. F. 1990; 7 (2): 289-293

    Abstract

    We have isolated and sequenced the gene encoding the human embryonic/atrial myosin alkali light chain isoform (MLC-1emb/A). The gene is split into seven exons by six introns; the last exon, as in all MLC isoform genes sequenced to date, is completely 3' untranslated sequence. Comparison of the MLC-1emb/A isoform gene with the other MLC-1 genes showed that the exon-intron arrangement of the human MLC-1emb/A isoform gene is analogous to that of the other MLC-1 type isoform genes. We have also mapped the human MLC-1emb/A isoform gene to the long arm of chromosome 17; the corresponding mouse gene has been mapped to chromosome 11. This gene, together with a number of others such as the collagen(I) alpha 1, galactokinase, and thymidine kinase genes, is part of the largest syntenic group between mouse and man.

    View details for Web of Science ID A1990DE81500021

    View details for PubMedID 2129532

  • A REGULAR PATTERN OF 2 TYPES OF 100-RESIDUE MOTIF IN THE SEQUENCE OF TITIN NATURE Labeit, S., BARLOW, D. P., Gautel, M., Gibson, T., Holt, J., Hsieh, C. L., FRANCKE, U., Leonard, K., WARDALE, J., Whiting, A., Trinick, J. 1990; 345 (6272): 273-276

    Abstract

    Titin is the largest polypeptide yet described (relative molecular mass approximately 3 x 10(6); refs 1, 2) and an abundant protein of striated muscle. Its molecules are string-like and in vivo span from the M to Z-lines. I-band regions of titin are thought to make elastic connections between the thick filament and the Z-line, thereby forming a third type of sarcomere filament. These would centre the A-band in the sarcomere and provide structural continuity in relaxed myofibrils. The A-band region of titin seems to be bound to the thick filament, where it has been proposed to act as a 'molecular ruler' regulating filament length and assembly. Here, we show that partial titin complementary DNAs encode a regular pattern of two types of 100-residue motif, each of which probably folds into a separate domain type. Such motifs are present in several evolutionarily divergent muscle proteins, all of which are likely to interact with myosin. One or both of the domain types is therefore likely to bind to myosin.

    View details for Web of Science ID A1990DD97300069

    View details for PubMedID 2129545

  • MOLECULAR ANALYSIS OF THE MOV-34 MUTATION - TRANSCRIPT DISRUPTED BY PROVIRAL INTEGRATION IN MICE IS CONSERVED IN DROSOPHILA DEVELOPMENT Gridley, T., Gray, D. A., ORRWEAVER, T., Soriano, P., Barton, D. E., FRANCKE, U., Jaenisch, R. 1990; 109 (1): 235-242

    Abstract

    The Mov 34 mutation is a recessive embryonic lethal mutation caused by retroviral integration in the murine germline. This integration disrupts a transcription unit that appears to encode a novel protein. The Mov 34 proviral integration is located on mouse chromosome 8 and the human homolog of this gene has been mapped to chromosome region 16q23-q24. An evolutionarily conserved syntenic relationship exists between this region of human chromosome 16 and a region of mouse chromosome 8 that also contains oligosyndactyly (Os), another recessive lethal mutation. Genetic studies have ruled out Os as residing at the same locus as the Mov 34 integration. The Mov 34 transcript is conserved in evolution, and a Drosophila homolog appears to encode a protein with 62% amino acid identity to the murine protein. In situ hybridization to Drosophila polytene chromosomes localizes the Drosophila homolog to 60B,C on chromosome 2. Several Drosophila lethal mutations also map to this region.

    View details for Web of Science ID A1990DF42600025

    View details for PubMedID 2209467

  • CDNA CLONING OF HUMAN OXYSTEROL-BINDING PROTEIN AND LOCALIZATION OF THE GENE TO HUMAN CHROMOSOME-11 AND MOUSE CHROMOSOME-19 GENOMICS Levanon, D., Hsieh, C. L., FRANCKE, U., Dawson, P. A., Ridgway, N. D., BROWN, M. S., Goldstein, J. L. 1990; 7 (1): 65-74

    Abstract

    Cellular cholesterol metabolism is regulated primarily through sterol-mediated feedback suppression of the activity of the low-density lipoprotein receptor and several enzymes of the cholesterol biosynthetic pathway. We previously described the cloning of a rabbit cDNA for the oxysterol-binding protein (OSBP), a cytosolic protein of 809 amino acids that may participate in these regulatory events. We now use the rabbit OSBP cDNA to clone the human OSBP cDNA and 5' genomic region. Comparison of the human and rabbit OSBP sequences revealed a remarkably high degree of conservation. The cDNA sequence in the coding region showed 94% identity between the two species, and the predicted amino acid sequence showed 98% identity. The human cDNA was used to determine the chromosomal localization of the OSBP gene by Southern blot hybridization to panels of somatic cell hybrid clones containing subsets of human or mouse chromosomes and by RFLP analysis of recombinant inbred mouse strains. The OSBP locus mapped to the long arm of human chromosome 11 and the proximal end of mouse chromosome 19. Along with previously mapped genes including Ly-1 and CD20, OSBP defines a new conserved syntenic group on the long arm of chromosome 11 in the human and the proximal end of chromosome 19 in the mouse.

    View details for Web of Science ID A1990DA11400008

    View details for PubMedID 1970801

  • THE GENE FOR THE UBIQUITOUS OCTAMER-BINDING PROTEIN OCT-1 IS ON HUMAN CHROMOSOME-1, REGION CEN-Q32, AND NEAR LY-22 AND LTW-4 ON MOUSE CHROMOSOME-1 GENOMICS Hsieh, C. L., Sturm, R., Herr, W., FRANCKE, U. 1990; 6 (4): 666-672

    Abstract

    Oct-1 is a sequence-specific transcription and DNA replication factor that recognizes the octameric sequence ATGCAAAT. This protein shares an extended region of sequence similarity, called the POU domain, with the lymphoid-specific transcription factor Oct-2, the pituitary-specific transcription factor Pit-1, and the Caenorhabditis elegans cell lineage gene product unc-86. Two subdomains, POU-related homeobox and POU-specific box, lie within the POU domain. Unlike other POU or homeodomain proteins, Oct-1 is ubiquitously expressed. A ubiquitous 95- to 100-kDa protein that has the same DNA-binding properties and is variously referred to as OTF-1, NFIII, or OBP100 is probably identical to Oct-1. The human gene was mapped by Southern blot analyses of human x rodent hybrid cell lines to chromosome 1, region cen-q32. In Chinese hamster x mouse hybrid cell lines the mouse locus was also mapped to chromosome 1. With the BXD (from progenitor strains C57BL/6J and DBA/2J) set of recombinant inbred strains of mice, the murine gene was localized on the linkage map of mouse chromosome 1. There were no recombinants among 26 strains with Ly-22 and 1 recombinant among 24 strains with Ltw-4. The gene symbol is OTF1 for humans and Oct-1 for mouse.

    View details for Web of Science ID A1990CW56300011

    View details for PubMedID 2341156

  • THE GENE FOR THE RNA COMPONENT OF THE MITOCHONDRIAL RNA-PROCESSING ENDORIBONUCLEASE IS LOCATED ON HUMAN CHROMOSOME-9P AND ON MOUSE CHROMOSOME-4 GENOMICS Hsieh, C. L., Donlon, T. A., DARRAS, B. T., Chang, D. D., Topper, J. N., Clayton, D. A., FRANCKE, U. 1990; 6 (3): 540-544

    Abstract

    Mitochondrial RNA-processing endoribonuclease (RNAase MRP) has the capacity to cleave mitochondrial RNA complementary to the light strand of the displacement loop at a unique site. The enzyme is a ribonucleoprotein whose RNA component is a nuclear gene product. The 5' flanking region of the primary transcript has control elements characteristic of RNA polymerase II transcription, and the coding region has features of RNA polymerase III transcription signals. The RNA associated with RNAase MRP is the first known RNA encoded by a single-copy gene in the nucleus and believed to be imported into mitochondria. The gene (RMRP) for this RNA component of RNAase MRP was assigned to human chromosome 9 and mouse chromosome 4 by Southern blot analyses of 11 human X rodent hybrids and 11 mouse X rodent hybrids with probe pHM1.0 and probe pSP270, respectively. In situ hybridization of probe pHSTU300 to normal human chromosomes revealed 29 of 100 cells with label on 9p and 9.6% of 302 silver grains located at 9p21--p12.

    View details for PubMedID 2328993

  • MOLECULAR-GENETICS - A GENE FOR WILMS TUMOR NATURE FRANCKE, U. 1990; 343 (6260): 692-694

    View details for Web of Science ID A1990CP80700026

    View details for PubMedID 2154698

  • NOVEL USE OF A CHIMPANZEE PSEUDOGENE FOR CHROMOSOMAL MAPPING OF HUMAN CYTOCHROME-C OXIDASE SUBUNIT-IV GENE Lomax, M. I., Welch, M. D., DARRAS, B. T., FRANCKE, U., Grossman, L. I. 1990; 86 (2): 209-216

    Abstract

    We have isolated a chimpanzee processed pseudogene for subunit IV of cytochrome c oxidase (COX; EC 1.9.3.1) by screening a chimpanzee genomic library in lambda Charon 32 with a bovine liver cDNA encoding COX subunit IV (COX IV), and localized it to a 1.9-kb HindIII fragment. Southern-blot analysis of genomic DNA from five primates showed that DNAs from human, gorilla, and chimpanzee each contained the 1.9-kb pseudogene fragment, whereas orangutan and pigtail macaque monkey DNA did not. This result clearly indicates that the pseudogene arose before the divergence of the chimpanzee and gorilla from the primate lineage. By screening Chinese hamster x human hybrid panels with the human COX4 cDNA, we have mapped COX4 genes to two human chromosomes, 14 and 16. The 1.9-kb HindIII fragment containing the pseudogene, COX4P1, can be assigned to chromosome 14, and by means of rearranged chromosomes in somatic cell hybrids, to 14q21-qter. Similarly, the functional gene, COX4, has been mapped to 16q22-qter.

    View details for Web of Science ID A1990CV70300010

    View details for PubMedID 2157630

  • CHROMOSOMAL ORGANIZATION OF ADRENERGIC-RECEPTOR GENES PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA YANGFENG, T. L., Xue, F. Y., Zhong, W. W., Cotecchia, S., Frielle, T., Caron, M. G., Lefkowitz, R. J., FRANCKE, U. 1990; 87 (4): 1516-1520

    Abstract

    The adrenergic receptors (ARs) (subtypes alpha 1, alpha 2, beta 1, and beta 2) are a prototypic family of guanine nucleotide binding regulatory protein-coupled receptors that mediate the physiological effects of the hormone epinephrine and the neurotransmitter norepinephrine. We have previously assigned the genes for beta 2- and alpha 2-AR to human chromosomes 5 and 10, respectively. By Southern analysis of somatic cell hybrids and in situ chromosomal hybridization, we have now mapped the alpha 1-AR gene to chromosome 5q32----q34, the same position as beta 2-AR, and the beta 1-AR gene to chromosome 10q24----q26, the region where alpha 2-AR is located. In mouse, both alpha 2- and beta 1-AR genes were assigned to chromosome 19, and the alpha 1-AR locus was localized to chromosome 11. Pulsed field gel electrophoresis has shown that the alpha 1- and beta 2-AR genes in humans are within 300 kilobases (kb) and the distance between the alpha 2- and beta 1-AR genes is less than 225 kb. The proximity of these two pairs of AR genes and the sequence similarity that exists among all the ARs strongly suggest that they are evolutionarily related. Moreover, they likely arose from a common ancestral receptor gene and subsequently diverged through gene duplication and chromosomal duplication to perform their distinctive roles in mediating the physiological effects of catecholamines. The AR genes thus provide a paradigm for understanding the evolution of such structurally conserved yet functionally divergent families of receptor molecules.

    View details for Web of Science ID A1990CP23800054

    View details for PubMedID 2154750

    View details for PubMedCentralID PMC53506

  • STRUCTURE AND CHROMOSOMAL MAPPING OF A HIGHLY POLYMORPHIC REPETITIVE DNA-SEQUENCE FROM THE PSEUDOAUTOSOMAL REGION OF THE MOUSE SEX-CHROMOSOMES CYTOGENETICS AND CELL GENETICS Harbers, K., FRANCKE, U., Soriano, P., Jaenisch, R., Muller, U. 1990; 53 (2-3): 129-133

    Abstract

    The pseudoautosomal region of the Mov15 mouse strain is marked by a Moloney murine leukemia provirus. The sequences flanking the Mov15 provirus were molecularly cloned and shown to consist of a tandemly repeated sequence of 31 nucleotides. Copy number variation of this repeat most likely accounts for the polymorphism in the mouse pseudoautosomal region detected with a probe from the flanking sequences. In situ hybridization to metaphase chromosomes showed heavy labeling of the pairing region of the X and Y chromosomes. The repetitive sequence was also found at the subtelomeric region of three autosomes. A similar level of amplification as the one seen on the sex chromosomes seems to be present on chromosomes 9 and 13. Lower copy number appear to be present on chromosome 4.

    View details for Web of Science ID A1990DL00300015

    View details for PubMedID 1973377

  • ISOLATION, CHROMOSOMAL MAPPING, AND EXPRESSION OF THE MOUSE TYROSINASE GENE JOURNAL OF INVESTIGATIVE DERMATOLOGY Kwon, B. S., Haq, A. K., Wakulchik, M., Kestler, D., Barton, D. E., FRANCKE, U., Lamoreux, M. L., Whitney, J. B., Halaban, R. 1989; 93 (5): 589-594

    Abstract

    Using a human tyrosinase cDNA probe, we have isolated mouse tyrosinase genomic clones and used them to map the mouse tyrosinase locus and to analyze the promoter sequence of the tyrosinase gene. Southern blot analyses of DNA from somatic cell hybrids, interspecies backcross mice, and albino deletion mice have revealed that the locus for mouse tyrosinase resides at or near the albino locus on mouse chromosome 7. There were three TATA-elements, but only one CAT-element, and the CAT-element appeared to be paired with the third TATA-element, located at the position farthest upstream. Mouse tyrosinase mRNA is approximately 2.4 Kb in size. The amount of tyrosinase mRNA reflects the levels of tyrosinase activity in normal melanocytes and Cloudman S-91 melanoma cell line.

    View details for Web of Science ID A1989AV87000001

    View details for PubMedID 2507645

  • CHROMOSOME LOCALIZATION AND CDNA SEQUENCE OF MURINE AND HUMAN GENES FOR RAS P21 GTPASE ACTIVATING PROTEIN (GAP) SOMATIC CELL AND MOLECULAR GENETICS Hsieh, C. L., VOGEL, U. S., Dixon, R. A., FRANCKE, U. 1989; 15 (6): 579-590

    Abstract

    The cDNA coding for mouse and human ras p21 GTPase-activating protein (GAP) was isolated; the deduced amino acid sequences share over 96% homology with that previously determined for bovine brain GAP. Both the mouse and human GAP cDNAs were used as probes for the chromosomal localization of this gene. The locus designations for the gene encoding GAP in human and mouse are RASA and Rasa (for ras-activating protein), respectively. By somatic cell hybrid analysis and in situ chromosomal hybridization, we have assigned the RASA gene to human chromosome band 5q13.3. In addition, with somatic cell genetics and linkage analysis in recombinant inbred mouse strains, the murine Rasa gene was localized to the distal end of mouse chromosome 13. These assignments place the gene encoding GAP in a known conserved syntenic region.

    View details for Web of Science ID A1989CG05300010

    View details for PubMedID 2574500

  • LEVEL OF EXPRESSION AND CHROMOSOME MAPPING OF THE MOUSE CHOLECYSTOKININ GENE - IMPLICATIONS FOR MURINE MODELS OF GENETIC OBESITY GENOMICS Friedman, J. M., Schneider, B. S., Barton, D. E., FRANCKE, U. 1989; 5 (3): 463-469

    Abstract

    Cholecystokinin (CCK) is a neuropeptide which is present in brain and intestine and which stimulates gall bladder contraction and pancreatic secretion. Additional studies have demonstrated an appetite-suppressing effect of CCK in vivo. These data have aroused speculation that the physiology of this hormone could be relevant in the pathogenesis of the mouse obesity mutations ob on chromosome 6 and db on chromosome 4. In order to determine whether abnormalities of this hormone could be the primary defect in these obesity mutations, we have used three separate approaches to map the mouse Cck gene to distal chromosome 9, where it is part of a syntenic group between mouse chromosome 9 and human chromosome 3. These data therefore exclude cholecystokinin as the etiologic factor in the pathogenesis of any of the known mouse obesity syndromes. In order to exclude the possibility that there are differences in mutant animals in the level of CCK RNA, we have used an S1 nuclease protection assay as well as a novel radioimmunoassay that detects the CCK precursor, to show that there are no gross differences in CCK mRNA or protein precursor levels between ob/ob and wild-type animals.

    View details for Web of Science ID A1989AT14200010

    View details for PubMedID 2575582

  • THE MOLECULAR-BASIS FOR DUCHENNE VERSUS BECKER MUSCULAR-DYSTROPHY - CORRELATION OF SEVERITY WITH TYPE OF DELETION AMERICAN JOURNAL OF HUMAN GENETICS Koenig, M., Beggs, A. H., Moyer, M., SCHERPF, S., HEINDRICH, K., Bettecken, T., Meng, G., Muller, C. R., Lindlof, M., Kaariainen, H., DELACHAPELLE, A., Kiuru, A., Savontaus, M. L., Gilgenkrantz, H., Recan, D., Chelly, J., KAPLAN, J. C., Covone, A. E., Archidiacono, N., Romeo, G., LIECHTIGALLATI, S., SCHNEIDER, V., Braga, S., Moser, H., DARRAS, B. T., Murphy, P., FRANCKE, U., Chen, J. D., Morgan, G., Denton, M., GREENBERG, C. R., WROGEMANN, K., BLONDEN, L. A., VANPAASSEN, H. M., VANOMMEN, G. J., KUNKEL, L. M. 1989; 45 (4): 498-506

    Abstract

    About 60% of both Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD) is due to deletions of the dystrophin gene. For cases with a deletion mutation, the "reading frame" hypothesis predicts that BMD patients produce a semifunctional, internally deleted dystrophin protein, whereas DMD patients produce a severely truncated protein that would be unstable. To test the validity of this theory, we analyzed 258 independent deletions at the DMD/BMD locus. The correlation between phenotype and type of deletion mutation is in agreement with the "reading frame" theory in 92% of cases and is of diagnostic and prognostic significance. The distribution and frequency of deletions spanning the entire locus suggests that many "in-frame" deletions of the dystrophin gene are not detected because the individuals bearing them are either asymptomatic or exhibit non-DMD/non-BMD clinical features.

    View details for Web of Science ID A1989AW39500003

    View details for PubMedID 2491009

  • GENE FOR ALPHA-2(I) COLLAGEN IS ON MOUSE CHROMOSOME-6 NOT 16 SOMATIC CELL AND MOLECULAR GENETICS Sundaresan, S., FRANCKE, U. 1989; 15 (5): 471-473

    Abstract

    Collagen is the most abundant protein in the animal world and a principal component, of the extracellular matrix of tissues. Type I collagen is composed of two alpha 1 chains and one alpha 2 chain. The human alpha 2(I) locus (COL1A2) has been assigned to human chromosome 7q21.3-q22.1. Here, we report the mapping of its murine counterpart Colla-2 to mouse chromosome 6 (MMU6) by Southern blotting using somatic cell hybrids. This result disagrees with the previously reported mapping of Colla-2 to MMU16 by immunochemical techniques. Our results are supported by comparative mapping data showing conserved homology between regions of human chromosome 7 and mouse chromosome 6.

    View details for Web of Science ID A1989AR39600010

    View details for PubMedID 2781418

  • GENES FOR HMG-COA REDUCTASE AND SEROTONIN-1A RECEPTOR ARE ON MOUSE CHROMOSOME-13 SOMATIC CELL AND MOLECULAR GENETICS Sundaresan, S., YANGFENG, T. L., FRANCKE, U. 1989; 15 (5): 465-469

    Abstract

    3-Hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase is the key regulatory enzyme for cholesterol biosynthesis. The human gene (HMGCR) has been assigned to the q13.3-q14 region of chromosome 5 (HSA5). We have now mapped the mouse gene Hmgcr to mouse chromosome 13 by Southern analysis of somatic cell hybrids. We also report the mapping to mouse chromosome 13 of the murine homolog of the gene for an intronless beta 2-adrenergic-like receptor, which is also located on human chromosome 5 region q11.2-q13 and has recently been identified as the serotonin 1a receptor. Our results confirm the existence of an evolutionarily conserved syntenic group of genes on the proximal long arm of HSA5 and on MMU13 that also includes the loci for arylsulfatase B, hexosaminidase B and dihydrofolate reductase.

    View details for Web of Science ID A1989AR39600009

    View details for PubMedID 2781417

  • ASSIGNMENT OF HUMAN GENES FOR PHOSPHORYLASE-KINASE SUBUNITS ALPHA-(PHKA) TO XQ12-Q13 AND BETA-(PHKB) TO 16Q12-Q13 AMERICAN JOURNAL OF HUMAN GENETICS FRANCKE, U., DARRAS, B. T., ZANDER, N. F., Kilimann, M. W. 1989; 45 (2): 276-282

    Abstract

    Phosphorylase kinase (PHK), the enzyme that activates glycogen phosphorylases in muscle, liver, and other tissues, is composed of four different subunits. Recently isolated rabbit muscle cDNAs for the larger two subunits, alpha and beta, have been used to map the location of their cognate sequences on human chromosomes. Southern blot analysis of rodent x human somatic cell hybrid panels, as well as in situ chromosomal hybridization, have provided evidence of single sites for both genes. The alpha subunit gene (PHKA) is located on the proximal long arm of the X chromosome in region Xq12-q13 near the locus for phosphoglycerate kinase (PGK1). X-linked mutations leading to PHK deficiency, known to exist in humans and mice, are likely to involve this locus. This hypothesis is consistent with the proximity of the Phk and Pgk-1 loci on the mouse X chromosome. In contrast, the beta subunit gene (PHKB) was found to be autosomal and was mapped to chromosome 16, region q12-q13 on the proximal long arm. Several different autosomally inherited forms of PHK deficiency for which the PHKB could be a candidate gene have been described in humans and rats.

    View details for Web of Science ID A1989AK38000010

    View details for PubMedID 2757032

  • BROTHER SISTER PAIRS AFFECTED WITH EARLY-ONSET, PROGRESSIVE MUSCULAR-DYSTROPHY - MOLECULAR STUDIES REVEAL ETIOLOGIC HETEROGENEITY AMERICAN JOURNAL OF HUMAN GENETICS FRANCKE, U., DARRAS, B. T., Hersh, J. H., Berg, B. O., MILLER, R. G. 1989; 45 (1): 63-72

    Abstract

    An autosomal recessive (AR) form of muscular dystrophy that clinically resembles Duchenne/Becker types exists, but its frequency is unknown. We have studied three unrelated affected brother/sister pairs and their families for deletions and polymorphisms with the entire dystrophin cDNA and other DNA probes from the Xp21 region to test for involvement of the DMD locus. In family 1 a large intragenic deletion was found in the affected male. The affected sister was heterozygous for this deletion, but the mother was not, implying germinal mosaicism. In family 2, no deletion was detected in the affected male. RFLP analysis revealed that the affected male and an unaffected sister shared a complete Xp21 haplotype while the affected sister had inherited a recombinant Xp21 region resulting from a crossover between pERT 87-15 and J-Bir. Only the 5' region of the dystrophin gene was shared with the affected boy. X-inactivation studies using a polymorphism in the 5'-flanking region of the HPRT gene, in conjunction with methylation-sensitive enzymes, revealed random X inactivation in the affected girl's leukocytes. In a muscle biopsy from the affected male, the dystrophin protein was present in normal amount and size. Family 3 was informative for four RFLPs detected with dystrophin cDNA probes which span the entire gene. The affected male was found to share the complete dystrophin RFLP haplotype with his unaffected brother, while his affected sister had inherited the other maternal haplotype. It is concluded that the clinical presentation of early-onset, progressive muscular dystrophy in a male and in his karyotypically normal sister can be caused by mutations at different loci. While in family 1 a deletion in the dystrophin gene is responsible, this gene does not appear to be involved in families 2 and 3.

    View details for Web of Science ID A1989AE77900008

    View details for PubMedID 2568091

  • INSULIN-LIKE GROWTH FACTOR-I RECEPTOR GENE IS CONCORDANT WITH C-FES PROTOONCOGENE AND MOUSE CHROMOSOME-7 IN SOMATIC-CELL HYBRIDS SOMATIC CELL AND MOLECULAR GENETICS Sundaresan, S., FRANCKE, U. 1989; 15 (4): 373-377

    Abstract

    The insulin-like growth factor I (IGF-1) mediates the actions of pituitary growth hormone in a variety of tissues. Its receptor (IGF1R) displays considerable structural similarity to the insulin receptor. In humans, the IGF1R gene has been mapped near FES, the cellular counterpart of the feline sarcoma virus transforming gene v-fes, at the q25-q26 region of human chromosome 15 (HSA15). Here, we report the mapping of mouse Igf1r to mouse chromosome 7 (MMU7) by somatic cell hybrid analysis. This result, along with the prior assignment of the loci for mitochondrial isocitrate dehydrogenase and FES to human chromosome 15 and mouse chromosome 7, suggest a conserved autosomal synteny group on the distal long arm of HSA15 and in the center of MMU7.

    View details for Web of Science ID A1989AL14500011

    View details for PubMedID 2548293

  • GENES FOR BETA-2-ADRENERGIC RECEPTOR AND PLATELET-DERIVED GROWTH-FACTOR RECEPTOR MAP TO MOUSE CHROMOSOME-18 SOMATIC CELL AND MOLECULAR GENETICS Sundaresan, S., FRANCKE, U. 1989; 15 (4): 367-371

    Abstract

    The beta 2-adrenergic receptor (ADRB2R) mediates the response of various cel types to neurotransmitters, hormones, and drugs. The platelet-derived growth factor (PDGF) interacts with its receptor (PDGFR) to stimulate mesenchymal cell proliferation. In the human, ADRB2R and PDGFR have been mapped to the q31--q32 region of chromosome 5 (HSA5). Here we report the mapping of Pdgfr and Adrb2r to mouse chromosome 18 (MMU18) using somatic cell hybrid mapping techniques. Together with previous mapping of genes for the glucocorticoid receptor (human locus GRL; mouse locus Gr1-1), the class II HLA invariant chain (human locus PHLAG; mouse locus Ii) and the FMS protooncogene to HSA5 and MMU18, the assignment of both Pdgfr and Adrb2r to MMU18 expands the conserved autosomal syntenic group.

    View details for Web of Science ID A1989AL14500010

    View details for PubMedID 2569767

  • MAPPING OF GENES FOR INHIBIN SUBUNIT-ALPHA, SUBUNIT-BETA-A, AND SUBUNIT-BETA-B ON HUMAN AND MOUSE CHROMOSOMES AND STUDIES OF JSD MICE GENOMICS Barton, D. E., YANGFENG, T. L., Mason, A. J., Seeburg, P. H., FRANCKE, U. 1989; 5 (1): 91-99

    Abstract

    Inhibin (INH) is a gonadal glycoprotein hormone that regulates pituitary FSH secretion and may also play a role in the regulation of androgen biosynthesis. There are two forms of inhibin that strongly inhibit pituitary FSH secretion. These share the same alpha subunit that is covalently linked to one of two distinct beta subunits (beta A or beta B). However, dimers of two beta subunits are potent stimulators of FSH synthesis and release in vitro. The beta subunits share extensive sequence similarity with transforming growth factor beta. Recently isolated cDNAs for all three inhibin subunits have been used to map their cognate loci on human and mouse chromosomes by Southern blot analysis of somatic cell hybrid DNAs and by in situ hybridization. INH alpha and INH beta B genes were assigned to human chromosome 2, regions q33----qter and cen----q13, respectively, and to mouse chromosome 1. The INH beta A locus was mapped to human chromosome 7p15----p14 and mouse chromosome 13. The region of mouse chromosome 1 that carries other genes known to have homologs on human chromosome 2q includes the jsd locus (for juvenile spermatogonial depletion). Adult jsd/jsd mice have elevated levels of serum FSH and their testes are devoid of spermatogonial cells. The possibility that the mutation in jsd involves the INH alpha or INH beta B gene was investigated by Southern blotting of DNA from jsd/jsd mice, and no major deletions or rearrangements were detected.

    View details for Web of Science ID A1989AB34200013

    View details for PubMedID 2767687

  • A GENE SPECIFYING SUBUNIT-VIII OF HUMAN CYTOCHROME-C OXIDASE IS LOCALIZED TO CHROMOSOME-11 AND IS EXPRESSED IN BOTH MUSCLE AND NON-MUSCLE TISSUES JOURNAL OF BIOLOGICAL CHEMISTRY Rizzuto, R., Nakase, H., Darras, B., FRANCKE, U., Fabrizi, G. M., MENGEL, T., Walsh, F., Kadenbach, B., DiMauro, S., Schon, E. A. 1989; 264 (18): 10595-10600

    Abstract

    Subunit VIII of mammalian cytochrome c oxidase (COX; EC 1.9.3.1) exists in at least two isoforms, because different but related polypeptides have been identified in COX isolated from liver and heart of both beef and pig. We have isolated a full length cDNA specifying subunit VIII of human COX from a human liver cDNA library. Sequences hybridizing to this cDNA are present at only one site, the COX8 locus, on human chromosome 11q12-q13. The deduced human polypeptide is 58% identical with COX VIII isolated from beef liver, but only 38% identical with COX VIII isolated from beef heart. Transcriptional analysis shows that an mRNA identical with the isolated cDNA is present in abundant amounts not only in human and monkey liver tissue, but in heart and skeletal muscle as well, tissues not known previously to contain this isoform. Since the only COX VIII subunit found in human heart agrees 100% with the polypeptide deduced from this coxVIII cDNA, it may well be that, in distinction to other mammals, only one form of COX VIII exists in primates.

    View details for Web of Science ID A1989AB64300043

    View details for PubMedID 2543673

  • CHROMOSOMAL LOCALIZATION OF THE GENE FOR THE HUMAN TRIFUNCTIONAL ENZYME, METHYLENETETRAHYDROFOLATE DEHYDROGENASE METHENYLTETRAHYDROFOLATE CYCLOHYDROLASE FORMYLTETRAHYDROFOLATE SYNTHETASE AMERICAN JOURNAL OF HUMAN GENETICS Rozen, R., Barton, D., Du, J., Hum, D. W., MacKenzie, R. E., FRANCKE, U. 1989; 44 (6): 781-786

    Abstract

    A trifunctional protein in man, 5,10-methylenetetrahydrofolate dehydrogenase-5,10-methenyltetrahydrofolate cyclohydrolase-10-formyltetrahydrofolate synthetase, catalyzes three consecutive steps in the interconversion of tetrahydrofolate derivatives; these derivatives supply one-carbon units for intermediary metabolism. Somatic cell hybridization and in situ hybridization were used to localize the functional gene coding for this protein--to human chromosome 14q24, near the c-fos and TGF-beta 3 loci. A second hybridizing sequence, possibly a pseudogene, was identified near the centromere of the X chromosome, at Xp11.

    View details for Web of Science ID A1989U854900001

    View details for PubMedID 2786332

  • A COMMON SACI POLYMORPHISM IN THE GENE FOR THE M1-SUBUNIT OF RIBONUCLEOTIDE REDUCTASE (RRM1) NUCLEIC ACIDS RESEARCH BATTAT, L., FRANCKE, U. 1989; 17 (10): 4005-4005

    View details for Web of Science ID A1989U846700059

    View details for PubMedID 2567505

  • NSI I RFLP AT THE X-LINKED CHRONIC GRANULOMATOUS-DISEASE LOCUS (CYBB) NUCLEIC ACIDS RESEARCH BATTAT, L., FRANCKE, U. 1989; 17 (9): 3619-3619

    View details for Web of Science ID A1989U545200052

    View details for PubMedID 2566976

  • THE GLUCOCORTICOID RECEPTOR GENE IS IN 5Q-Q32 GENOMICS FRANCKE, U., FOELLMER, B. E. 1989; 4 (4): 610-612

    View details for Web of Science ID A1989U455000020

    View details for PubMedID 2744768

  • 2 RFLPS ASSOCIATED WITH THE HUMAN ENDOGENOUS RETROVIRAL ELEMENT-S71 ON CHROMOSOME-18Q21 NUCLEIC ACIDS RESEARCH LEIBMOSCH, C., Barton, D., Geigl, E. M., BRACKWERNER, R., Erfle, V., Hehlmann, R., FRANCKE, U. 1989; 17 (6): 2367-2367

    View details for Web of Science ID A1989T902200030

    View details for PubMedID 2565025

  • BLAST-1 POSSESSES A GLYCOSYL-PHOSPHATIDYLINOSITOL (GPI) MEMBRANE ANCHOR, IS RELATED TO LFA-3 AND OX-45, AND MAPS TO CHROMOSOME-1Q21-23 JOURNAL OF EXPERIMENTAL MEDICINE Staunton, D. E., Fisher, R. C., LEBEAU, M. M., Lawrence, J. B., Barton, D. E., FRANCKE, U., Dustin, M., THORLEYLAWSON, D. A. 1989; 169 (3): 1087-1099

    Abstract

    Blast-1 is a human activation-associated glycoprotein expressed on the surface of leukocytes. Analysis of a translated sequence from a Blast-1 cDNA reveals a single hydrophobic sequence which could traverse the plasma membrane, but is devoid of charged residues that might represent a cytoplasmic tail. Consistent with this characteristic, Blast-1 is demonstrated here to be anchored to the cell surface through a glycosyl-phosphatidylinositol (GPI)-containing lipid. Comparison of Blast-1 to other GPI-anchored membrane proteins revealed a striking primary and secondary structure similarity with MRC OX45 and the lymphocyte function antigen LFA-3. The degree of overall amino acid sequence homology reveals that OX45 is a rat homologue of Blast-1. The greatest homology to LFA-3 occurs between their NH2-terminal Ig-like domains. Evidence is presented that demonstrates that Blast-1 and LFA-3 possess a disulfide-bonded second domain. These common characteristics demonstrate a structural and evolutionary relationship between Blast-1, OX45, LFA-3, and CD2, which in turn suggests a functional role for Blast-1 in cell-cell interactions in the immune response. The gene for Blast-1 has been localized to chromosome 1 q21-q23, indistinguishable from the CD1 cluster of Ig superfamily genes, raising the possibility that they may be linked.

    View details for Web of Science ID A1989T533800036

    View details for PubMedID 2466936

  • HUMAN VENTRICULAR SLOW TWITCH MYOSIN ALKALI LIGHT CHAIN GENE CHARACTERIZATION, SEQUENCE, AND CHROMOSOMAL LOCATION JOURNAL OF BIOLOGICAL CHEMISTRY Fodor, W. L., Darras, B., Seharaseyon, J., FALKENTHAL, S., FRANCKE, U., Vanin, E. F. 1989; 264 (4): 2143-2149

    Abstract

    The gene coding for the human ventricular/slow twitch myosin alkali light chain isoform was isolated and sequenced. It was found to contain a total of seven exons, the last of which is completely 3'-untranslated sequence. Comparison of this gene sequence with that of the various fast twitch skeletal isoform gene sequences revealed that the exon-intron arrangement is conserved within the myosin alkali light chain gene family. In fact the introns are in exactly the same positions within analogous codons. Comparison of the derived amino acid sequence from the human ventricular/slow twitch isoform gene with that of other isoform protein sequences indicated that the protein encoded by this gene is more homologous to the chicken cardiac isoform protein sequence than to any of the other protein sequences. These results indicate that the gene duplication which gave rise to the ventricular/slow twitch and fast twitch isoform genes must have occurred prior to the divergence of mammals and avians. We have also localized the human ventricular/slow twitch isoform gene to the short arm of human chromosome 3. Interestingly the corresponding mouse gene has been mapped to the distal region of mouse chromosome 9 which contains a conserved syntenic group of genes that map to the short arm of human chromosome 3.

    View details for Web of Science ID A1989T106200043

    View details for PubMedID 2789520

  • DYSTROPHIN IMMUNOCYTOCHEMISTRY IN MUSCLE CULTURE - DETECTION OF A CARRIER OF DUCHENNE MUSCULAR-DYSTROPHY AMERICAN JOURNAL OF MEDICAL GENETICS Miranda, A. F., FRANCKE, U., Bonilla, E., Martucci, G., Schmidt, B., Salviati, G., Rubin, M. 1989; 32 (2): 268-273

    Abstract

    Dystrophin is the gene product which is affected in Duchenne muscular dystrophy (DMD). We studied differentiating clonal muscle cultures derived from normal muscle and from the mother of a DMD patient by immunocytochemistry, using anti-dystrophin antibody. While clonal cultures derived from normal muscle expressed dystrophin in all myotubes, two populations of myogenic cells could be demonstrated in muscle from this possible DMD carrier; in 13 clones the myotubes expressed dystrophin and in 7 clones dystrophin was undetectable. No DNA deletion, duplication or rearrangement was detected by Southern blot analysis of DNA from this family using cDNA probes. Thus, immunocytochemical analysis of clonal muscle cultures may be a useful method to determine whether mothers of DMD patients are carriers of the DMD mutation, especially in the absence of demonstrable gene defects.

    View details for Web of Science ID A1989T365800077

    View details for PubMedID 2648829

  • ADRENAL GLUCOCORTICOIDS REGULATE ADIPSIN GENE-EXPRESSION IN GENETICALLY-OBESE MICE JOURNAL OF BIOLOGICAL CHEMISTRY Spiegelman, B. M., Lowell, B., Napolitano, A., DUBUC, P., Barton, D., FRANCKE, U., GROVES, D. L., Cook, K. S., Flier, J. S. 1989; 264 (3): 1811-1815

    Abstract

    Adipsin expression at the protein and mRNA levels is greatly reduced in several distinct syndromes of obesity in the mouse: genetic obesity due to the db/db and ob/ob genes, and a chemically induced model secondary to neonatal exposure to monosodium glutamate. We considered first the possibility that the adipsin gene might be identical to the db or ob locus and the lowered expression of this protein might result from a mutation in this gene. We show here that the adipsin structural gene is located on chromosome 10 and hence is physically distinct from any obesity genes so far identified in the mouse. A major role for the adrenal gland and adrenal glucocorticoids in the aberrant regulation of adipsin in these models of obesity is indicated by several experiments. Adrenalectomy of the ob/ob mouse raises the circulating levels of adipsin protein and the amount of this mRNA in epididymal fat pads (5-fold), although neither is increased to the levels seen in lean controls. Exogenous administration of corticosterone completely blocks the effects of adrenalectomy on adipsin, suggesting that the effect of this endocrine ablation is through reduction of adrenal glucocorticoids. Corticosterone administration also causes suppression in the levels of adipsin mRNA and protein in lean mice, although this decrease is never as severe as that seen in obese mice. The effect of exogenous corticosterone in lean mice occurs within 2 days and hence is not secondary to the obesity which these hormones eventually elicit. These results indicate that glucocorticoids can regulate adipsin expression in vivo and strongly suggest that the hyperglucocorticoid state seen in certain obese models plays a significant role in lowering adipsin mRNA and protein levels. Quantitative analysis of these experiments suggests that other as yet unknown neuroendocrine factors also function to suppress adipsin in obesity.

    View details for Web of Science ID A1989R890300070

    View details for PubMedID 2912985

  • Genetic characterization of a human endogenous retroviral element located on chromosome 18q21. Haematology and blood transfusion Leib-Mösch, C., Barton, D. E., Brack-Werner, R., FOELLMER, B., Werner, T., Rohrmeier, D., FRANCKE, U., Erfle, V., Hehlmann, R. 1989; 32: 461-463

    View details for PubMedID 2576240

  • THE GENES FOR 2 NEUTROPHIL CYTOSOL FACTORS THAT ARE DEFICIENT IN AUTOSOMAL FORMS OF CHRONIC GRANULOMATOUS-DISEASE ARE ON HUMAN CHROMOSOME-10 (47-KD), AND CHROMOSOME-1 CEN-Q32 (65-KD) Hsieh, C. L., Leto, T. L., LOMAX, K. J., Malech, H. L., FRANCKE, U. KARGER. 1989: 1015–16
  • THE GENE FOR GTPASE ACTIVATING PROTEIN (GAP) IS ON HUMAN CHROMOSOME-5Q AND MOUSE CHROMOSOME-13 Hsieh, C. L., FRANCKE, U. KARGER. 1989: 1016–16
  • CHROMOSOME MAPPING OF THE GROWTH-HORMONE RECEPTOR GENE IN MAN AND MOUSE CYTOGENETICS AND CELL GENETICS Barton, D. E., FOELLMER, B. E., Wood, W. I., FRANCKE, U. 1989; 50 (2-3): 137-141

    Abstract

    Pituitary growth hormone (GH) is essential for normal growth and development in animals and GH deficiency leads to dwarfism. This hormone acts via specific high-affinity cell surface receptors found in liver and other tissues. The recent cloning and sequencing of cDNAs encoding human and rabbit GH receptors (GHR) has demonstrated that this receptor is unrelated to any previously described cell membrane receptor or growth factor receptor. We have used the cloned human GHR cDNA to map the GHR locus to the proximal short arm of human chromosome 5, region p13.1----p12, and to mouse chromosome 15 by Southern blot analysis and in situ hybridization. While human chromosome 5 carries several genes for hormone and growth factor receptors, GHR is the only growth-related gene so far mapped to the short arm. Inasmuch as GHR is the first gene with apparently homologous loci on human chromosome 5 and mouse chromosome 15, it identifies a new homologous conserved region. In humans, deficiency of GH receptor activity probably causes Laron-type dwarfism, an autosomal recessive disorder prevalent in Oriental Jews. In mice, the autosomal recessive mutation miniature (mn) is characterized by severe growth failure and early death and has been mapped to chromosome 15. Our assignment of Ghr to mouse chromosome 15 suggests this as a candidate gene for the mn mutation.

    View details for Web of Science ID A1989AR04200018

    View details for PubMedID 2776481

  • HUMAN SSAV-RELATED ENDOGENOUS RETROVIRAL ELEMENT - LTR-LIKE SEQUENCE AND CHROMOSOMAL LOCALIZATION TO 18Q21 GENOMICS BRACKWERNER, R., Barton, D. E., Werner, T., FOELLMER, B. E., LEIBMOSCH, C., FRANCKE, U., Erfle, V., Hehlmann, R. 1989; 4 (1): 68-75

    Abstract

    A new family of human endogenous retroviral sequences was recently discovered by way of its relationship to the simian sarcoma-associated virus (SSAV). One molecular clone, termed S71, contains sequences related to the genes coding for the group-specific antigens (gag) and polymerase (pol) proteins of SSAV. At the 3' end of this human retroviral element we have now found a 535-bp region which shows features characteristics of a retroviral long terminal repeat, including potential signal sequences essential for transcriptional control. By means of Southern blotting and in situ hybridization, the sequence was mapped to chromosome 18 band q21.

    View details for Web of Science ID A1989R510100011

    View details for PubMedID 2536635

  • CHROMOSOME LOCALIZATION AND EXPRESSION PATTERN OF LMYC AND BMYC IN MURINE EMBRYONAL CARCINOMA-CELLS ONCOGENE Ingvarsson, S., Sundaresan, S., Jin, P., FRANCKE, U., Asker, C., SUMEGI, J., Klein, G., Sejersen, T. 1988; 3 (6): 679-685

    Abstract

    Using Southern blot analysis of DNA from mouse-hamster somatic cell hybrids, we have mapped Lmyc and Bmyc, two members of the myc family of genes, to mouse chromosomes 4 and 2, respectively. Furthermore, we have compared the regulation of Lmyc and Bmyc expression under different growth conditions and during in vitro differentiation of the murine EC line F9 and considered the findings in relation to our previous studies on Nmyc and c-myc expression in the same line (Sejersen et al., 1987). Lmyc was down-regulated at an early stage of visceral endoderm differentiation, similarly to c-myc and Nmyc, while Bmyc was expressed at a constant low level at all stages. Lmyc, but not c-myc and Nmyc, was upregulated in terminally differentiated visceral endoderm cells. Inhibition of protein synthesis by cycloheximide for 4 h induced a 70% increase in Lmyc and 30% increase in Bmyc transcript levels, indicating that the expression of these genes is negatively regulated by a short-lived protein. Mitogenic stimulation with insulin and transferrin did not affect Lmyc and Bmyc mRNA levels. Lmyc transcripts have a half life of 30 min, whereas the Bmyc transcript is highly stable, with a half life of 6 h. The half-lives of the c-myc and Nmyc transcripts have been estimated previously as 40 and 130 min, respectively.

    View details for Web of Science ID A1988R646000009

    View details for PubMedID 2577870

  • INTRAGENIC DELETIONS IN 21 DUCHENNE MUSCULAR-DYSTROPHY (DMD) BECKER MUSCULAR-DYSTROPHY (BMD) FAMILIES STUDIED WITH THE DYSTROPHIN CDNA - LOCATION OF BREAKPOINTS ON HINDIII AND BGIII EXON-CONTAINING FRAGMENT MAPS, MEIOTIC AND MITOTIC ORIGIN OF THE MUTATIONS AMERICAN JOURNAL OF HUMAN GENETICS DARRAS, B. T., Blattner, P., HARPER, J. F., Spiro, A. J., Alter, S., FRANCKE, U. 1988; 43 (5): 620-629

    Abstract

    Following the strategy outlined in an accompanying paper, we studied 32 X-linked muscular dystrophy families (29 Duchenne [DMD] and three Becker [BMD] type) for abnormalities of HindIII and BglII fragments detected by the entire dystrophin cDNA. Twenty-one different single-intragenic deletions, and no duplications, were identified. The deletion endpoints were precisely mapped on the published HindIII fragment map. Detailed analysis of overlapping deletions led to clarification of the fragment order for some previously unsettled regions of the HindIII map and to the construction of a partial map of exon-containing BglII fragments. For the regions involved in deletions, the corresponding HindIII and BglIII fragments could be identified. Noncontiguous comigrating fragments were detected in two regions by careful analysis of the patterns in deletion patients. Four of the 21 deletions generated novel restriction fragments that facilitated detection of female carriers in these families. Twelve of the deletions had a breakpoint in one of the two large introns known to be the sites of breakpoint clusters. By combining deletions and RFLP analyses, we unequivocally identified the gamete that first carried the mutation in 13 families: eight oocytes and five sperm. Germ-line mosaicism previously detected in one male was confirmed by cDNA studies. In two additional families gonadal mosaicism was found in females. As evidence is accumulating for frequent mitotic origin of these deletion mutations, this phenomenon has to be considered when postulating mutational mechanisms and in genetic counseling of DMD/BMD families.

    View details for Web of Science ID A1988Q933600008

    View details for PubMedID 2903663

  • NORMAL HUMAN GENOMIC RESTRICTION-FRAGMENT PATTERNS AND POLYMORPHISMS REVEALED BY HYBRIDIZATION WITH THE ENTIRE DYSTROPHIN CDNA AMERICAN JOURNAL OF HUMAN GENETICS DARRAS, B. T., FRANCKE, U. 1988; 43 (5): 612-619

    Abstract

    Since the complete cDNA for the gene that causes X-linked recessive Duchenne/Becker muscular dystrophy (DMD/BMD) when mutated or deleted has recently been cloned and made generally available, DNA-based diagnostic studies of affected males and their families have entered into a new era. This communication sets forth the standard patterns of restriction fragments that are detected when normal human DNA cleaved with either HindIII or BglII is hybridized with seven contiguous segments comprising the entire 14-kb cDNA. Collectively, the more than 60 restriction fragments allow visualization of approximately 350 (HindIII) to 400 (BglII) kbp. This corresponds to the exon-containing one-fifth of the total genomic length of this gene, including the 3' untranslated region. Twelve two-allele restriction-site polymorphisms that span the entire length of the gene were detected with the cDNA probes and allele frequencies determined. A diagnostic approach is proposed that starts with deletion screening of DNA from male probands, includes carrier detection based on relative fragment intensities, and extends to RFLP detection using the same autoradiographs prepared for deletion screening. Our results on deletion analysis of 32 DMD/BMD families are presented in an accompanying paper.

    View details for Web of Science ID A1988Q933600007

    View details for PubMedID 2903662

  • CHROMOSOMAL MAPPING OF THE GENE FOR THE TYPE-II INSULIN-LIKE GROWTH-FACTOR RECEPTOR CATION-INDEPENDENT MANNOSE 6-PHOSPHATE RECEPTOR IN MAN AND MOUSE GENOMICS Laureys, G., Barton, D. E., Ullrich, A., FRANCKE, U. 1988; 3 (3): 224-229

    Abstract

    The gene for insulin-like growth factor II (IGF-II) receptor (IGF2R) that has recently been found, by DNA sequencing, to be identical to the cation-independent mannose 6-phosphate receptor (CIM6PR) has been mapped in the human and murine species. Cloned cDNAs for human and rat IGF-II receptors were used to probe Southern blots of somatic cell hybrid DNA and for in situ chromosomal hybridization. The genes are located in a region of other conserved syntenic genes on the long arm of human chromosome 6, region 6q25----q27, and mouse chromosome 17, region A-C. The CIM6PR/IGF2R locus in man is asyntenic with the genes encoding IGF-II (IGF2), the IGF-I receptor (IGF1R), and the cation-dependent mannose 6-phosphate receptor (CDM6PR).

    View details for Web of Science ID A1988R066400007

    View details for PubMedID 2852162

  • CYTOGENETIC ANALYSIS OF MELANOCYTES FROM PREMALIGNANT NEVI AND MELANOMAS JOURNAL OF THE NATIONAL CANCER INSTITUTE Cowan, J. M., Halaban, R., FRANCKE, U. 1988; 80 (14): 1159-1164

    Abstract

    We karyotypically analyzed cultured melanocytes from a variety of lesions, including congenital and dysplastic nevi, primary melanoma, and metastatic melanoma. The cells derived from congenital nevi had normal karyotypes, as did 22 of the 26 cultures derived from dysplastic nevi. The karyotypes of melanocytes from primary and metastatic melanomas were all abnormal. The only chromosome change in common between the nevi with abnormal karyotypes and the melanomas was the loss of one copy of chromosome 9 (two of four nevi and four of 11 melanomas, including three from the same patient) or the loss of the short arm of chromosome 9, especially of region 9pter-p22 (three of 11 melanomas). We suggest that deletion of a gene or genes on 9p, possibly interferon genes, is an initial step in the malignant transformation of melanocytes.

    View details for Web of Science ID A1988Q102900015

    View details for PubMedID 3166071

  • CONGENITAL ADRENAL HYPOPLASIA AND SELECTIVE ABSENCE OF PITUITARY LUTEINIZING-HORMONE - A NEW AUTOSOMAL RECESSIVE SYNDROME AMERICAN JOURNAL OF MEDICAL GENETICS Burke, B. A., WICK, M. R., King, R., Thompson, T., Hansen, J., DARRAE, B. T., FRANCKE, U., Seltzer, W. K., McCabe, E. R., Scheithauer, B. W. 1988; 31 (1): 75-97

    Abstract

    Congenital hypoplasia of the adrenal glands (CHA) is a rare condition, particularly in the absence of a central nervous system (CNS) anomaly. Two major types of CHA have been described in the setting of an apparently normal CNS and pituitary: a cytomegalic type usually with X-linked recessive inheritance and a miniature adult type that, when hereditary, is an autosomal recessive trait. Glycerol kinase deficiency (GKD) is an X-linked recessive trait, and it may be associated with CHA and adrenal insufficiency, presumably because of deletion of adjacent X-linked loci. We report on three sibling infants, one male and two females, with normal CNS and lethal CHA of the miniature adult type, selective absence of pituitary LH; two of the infants also had glycerol kinase (GK) activity that was decreased but not in the GKD range. Restriction fragment length polymorphism (RFLP) analysis of X chromosome markers located at Xp21-p22 was carried out on the maternal grandfather, both parents, two of three affected infants, and a living normal brother. The results excluded the X-linked type of this disorder associated with GKD in this family. Autosomal recessive inheritance is most likely.

    View details for Web of Science ID A1988Q237500010

    View details for PubMedID 2906226

  • MYOPATHY IN COMPLEX GLYCEROL KINASE-DEFICIENCY PATIENTS IS DUE TO 3' DELETIONS OF THE DYSTROPHIN GENE AMERICAN JOURNAL OF HUMAN GENETICS DARRAS, B. T., FRANCKE, U. 1988; 43 (2): 126-130

    Abstract

    DNA samples from nine previously reported patients with X-linked recessive glycerol kinase deficiency, associated in seven of them with adrenal hypoplasia and in five with developmental delay and myopathy, have been studied for deletions of the Duchenne/Becker muscular dystrophy gene by probing with the entire cDNA for the dystrophin protein. All five patients with myopathy, including two in whom no deletions had been detected before, were found to have variable-sized deletions extending through the 3' end of this gene. The 5' deletion breakpoints are intragenic in four cases and have been mapped precisely on the exon-containing HindIII fragment map. A correlation was found between severity and progression of the muscular dystrophy phenotype and the sizes of the gene deletions. In cases in which there was glycerol kinase deficiency/adrenal hypoplasia microdeletion syndrome without myopathy, no deletions were found with the dystrophin cDNA.

    View details for Web of Science ID A1988P608400003

    View details for PubMedID 2840818

  • MOLECULAR CHARACTERIZATION AND CHROMOSOMAL MAPPING OF MELANOMA GROWTH STIMULATORY ACTIVITY, A GROWTH-FACTOR STRUCTURALLY RELATED TO BETA-THROMBOGLOBULIN EMBO JOURNAL Richmond, A., Balentien, E., Thomas, H. G., Flaggs, G., Barton, D. E., Spiess, J., Bordoni, R., FRANCKE, U., DERYNCK, R. 1988; 7 (7): 2025-2033

    Abstract

    Melanoma growth stimulatory activity (MGSA) is a mitogenic polypeptide secreted by Hs294T human melanoma cells. Comparison of the N-terminal sequences of the 13 and 16 kd MGSA species with the cDNA sequence revealed that the mature form of human MGSA is maximally 73 amino acids long. Expression of the cDNA in mammalian cells results in the secretion of this peptide with mitogenic activity. MGSA is structurally related to the platelet-derived beta-thromboglobulin and to several other polypeptides. These factors may constitute a family of growth factors. MGSA mRNA was detected in a variety of cell types. The level of MGSA mRNA in melanoma cells is strongly elevated by treatment with MGSA. MGSA is the gene product of a recently detected gene gro. The gene was mapped to chromosome 4 (region q13----q21). This same region also contains genes for two of the structurally related factors, for c-kit, a receptor for an as yet unidentified ligand, and for 'piebald trait', an inherited skin pigmentation disorder.

    View details for Web of Science ID A1988P074100014

    View details for PubMedID 2970963

  • A NOVEL PROTEIN TYROSINE KINASE GENE WHOSE EXPRESSION IS MODULATED DURING ENDOTHELIAL-CELL DIFFERENTIATION ONCOGENE Ruta, M., HOWK, R., Ricca, G., Drohan, W., Zabelshansky, M., Laureys, G., Barton, D. E., FRANCKE, U., Schlessinger, J., Givol, D. 1988; 3 (1): 9-15
  • Human tyrosinase gene, mapped to chromosome 11 (q14----q21), defines second region of homology with mouse chromosome 7. Genomics Barton, D. E., Kwon, B. S., FRANCKE, U. 1988; 3 (1): 17-24

    Abstract

    The enzyme tyrosinase (monophenol,L-dopa:oxygen oxidoreductase; EC 1.14.18.1) catalyzes the first two steps in the conversion of tyrosine to melanin, the major pigment found in melanocytes. Some forms of oculocutaneous albinism, characterized by the absence of melanin in skin and eyes and by a deficiency of tyrosinase activity, may result from mutations in the tyrosinase structural gene. A recently isolated human tyrosinase cDNA was used to map the human tyrosinase locus (TYR) to chromosome 11, region q14----q21, by Southern blot analysis of somatic cell hybrid DNA and by in situ chromosomal hybridization. A second site of tyrosinase-related sequences was detected on the short arm of chromosome 11 near the centromere (p11.2----cen). Furthermore, we have confirmed the localization of the tyrosinase gene in the mouse at or near the c locus on chromosome 7. Comparison of the genetic maps of human chromosome 11 and mouse chromosome 7 leads to hypotheses regarding the evolution of human chromosome 11.

    View details for PubMedID 3146546

  • THE GENE FOR CYSTATHIONINE BETA-SYNTHASE (CBS) MAPS TO THE SUBTELOMERIC REGION ON HUMAN CHROMOSOME-21Q AND TO PROXIMAL MOUSE CHROMOSOME-17 AMERICAN JOURNAL OF HUMAN GENETICS MUNKE, M., Kraus, J. P., Ohura, T., FRANCKE, U. 1988; 42 (4): 550-559

    Abstract

    The human gene for cystathionine beta-synthase (CBS), the enzyme deficient in classical homocystinuria, has been assigned to the subtelomeric region of band 21q22.3 by in situ hybridization of a rat cDNA probe to structurally rearranged chromosomes 21. The homologous locus in the mouse (Cbs) was mapped to the proximal half of mouse chromosome 17 by Southern analysis of Chinese hamster X mouse somatic cell hybrid DNA. Thus, CBS/Cbs and the gene for alpha A-crystalline (CRYA1/Crya-1 or Acry-1) form a conserved linkage group on human (HSA) chromosome region 21q22.3 and mouse (MMU) chromosome 17 region A-C. Features of Down syndrome (DS) caused by three copies of these genes should not be present in mice trisomic for MMU 16 that have been proposed as animal models for DS. Mice partially trisomic for MMU 16 or MMU 17 should allow gene-specific dissection of the trisomy 21 phenotype.

    View details for Web of Science ID A1988M631600004

    View details for PubMedID 2894761

  • REGIONAL ASSIGNMENT OF 6 POLYMORPHIC DNA-SEQUENCES ON CHROMOSOME-21 BY INSITU HYBRIDIZATION TO NORMAL AND REARRANGED CHROMOSOMES AMERICAN JOURNAL OF HUMAN GENETICS MUNKE, M., FOELLMER, B., WATKINS, P. C., Cowan, J. M., Carroll, A. J., Gusella, J. F., FRANCKE, U. 1988; 42 (4): 542-549

    Abstract

    We have assigned six polymorphic DNA segments to chromosomal subregions and have established the physical order of these sequences on the long arm of chromosome 21 by in situ hybridization of cloned probes to normal metaphase chromosomes and chromosomes 21 from individuals with three different structural rearrangements: an interstitial deletion, a ring chromosome, and a reciprocal translocation involving four different breakpoints in band 21q22. Segments D21S1 and D21S11 map to region 21q11.2----q21, D21S8 to 21q21.1----q22.11, and D21S54 to 21q21.3----q22.11; D21S23 and D21S25 are both in the terminal subband 21q22.3, but they are separated by a chromosomal breakpoint in a ring 21 chromosome, a finding that places D21S23 proximal to D21S25. The physical map order D21S1/D21S11-D21S8-D21S54-D21S23-D21S25 agrees with the linkage map, but genetic distances are disproportionately larger toward the distal end of 21q.

    View details for Web of Science ID A1988M631600003

    View details for PubMedID 3348217

  • DIRECT METHOD FOR PRENATAL-DIAGNOSIS AND CARRIER DETECTION IN DUCHENNE BECKER MUSCULAR-DYSTROPHY USING THE ENTIRE DYSTROPHIN CDNA AMERICAN JOURNAL OF MEDICAL GENETICS DARRAS, B. T., Koenig, M., KUNKEL, L. M., FRANCKE, U. 1988; 29 (3): 713-726

    Abstract

    DNA sequence polymorphisms (RFLPs) have been widely used as genetic markers for identification of the X chromosome that carries the mutation for Duchenne muscular dystrophy (DMD) in affected families, but serious limitations and pitfalls are associated with this approach [Darras et al., 1987]. The complementary DNA (cDNA) of the DMD gene has recently been isolated and shown to detect partial gene deletions in a large proportion of patients [Koenig et al., 1987]. Two prenatal studies are presented to illustrate how the unambiguous identification of deletion mutations by cDNA probes permits direct DNA-based diagnoses with high accuracy and in otherwise uninformative families. In a single proband family, DNA marker analysis had determined that the Xp21 chromosomal region present in the affected male was also carried by a male fetus in a subsequent pregnancy. Analysis of this family's DNA with probes covering the entire 14 kb cDNA revealed a small deletion in the affected male that was not present in the fetus nor in the mother. In the second family the fetus was a female deletion carrier identified by comparing intensities of restriction fragments. Since 1/3 of all DMD patients are thought to result from new mutations and most families have only single affected males, the cloned cDNA probes now available are likely to revolutionize DNA-based diagnostic studies in this disorder. More reliable, more rapid and less expensive than linkage studies with DNA polymorphisms, this method will be informative in the more than 50% of DMD/BMD cases that have deletion mutations.

    View details for Web of Science ID A1988M799200039

    View details for PubMedID 2897793

  • Chromosomal localization of human Na+, K+-ATPase alpha- and beta-subunit genes. Genomics Yang-Feng, T. L., Schneider, J. W., Lindgren, V., SHULL, M. M., Benz, E. J., Lingrel, J. B., FRANCKE, U. 1988; 2 (2): 128-138

    Abstract

    Na+, K+-ATPase is a heterodimeric enzyme responsible for the active maintenance of sodium and potassium gradients across the plasma membrane. Recently, cDNAs for several tissue-specific isoforms of the larger catalytic alpha-subunit and the smaller beta-subunit have been cloned. We have hybridized rat brain and human kidney cDNA probes, as well as human genomic isoform-specific DNA fragments, to Southern filters containing panels of rodent X human somatic cell hybrid lines. The results obtained have allowed us to assign the loci for the ubiquitously expressed alpha-chain (ATP1A1) to human chromosome 1, region 1p21----cen, and for the alpha 2 isoform that predominates in neural and muscle tissues (ATP1A2) to chromosome 1, region cen----q32. A common PstI RFLP was detected with the ATP1A2 probe. The alpha 3 gene, which is expressed primarily in neural tissues (ATP1A3), was assigned to human chromosome 19. A fourth alpha gene of unknown function (alpha D) that was isolated by molecular cloning (ATP1AL1) was mapped to chromosome 13. Although evidence to date had suggested a single gene for the beta-subunit, we found hybridizing restriction fragments derived from two different human chromosomes. On the basis of knowledge of conserved linkage groups on human and murine chromosomes, we propose that the coding gene ATP 1B is located on the long arm of human chromosome 1 and that the sequence on human chromosome 4 (ATP 1BL1) is either a related gene or a pseudogene.

    View details for PubMedID 2842249

  • ASSIGNMENT OF THE T-CELL DIFFERENTIATION GENE MAL TO HUMAN CHROMOSOME-2, REGION CEN-]Q13 IMMUNOGENETICS Alonso, M. A., Barton, D. E., FRANCKE, U. 1988; 27 (2): 91-95

    Abstract

    A cDNA clone has recently been isolated that encodes a protein expressed only in the intermediate and late stages of T-cell differentiation, termed MAL. The polypeptide deduced from the MAL cDNA sequence contains four potential transmembrane domains. We have used a panel of 28 human X rodent hybrid cell lines to assign the MAL gene to the proximal long arm of human chromosome 2, region cen----q13. The significance of the MAL map position and its relationship with other genes on chromosome 2 are discussed.

    View details for Web of Science ID A1988L332400003

    View details for PubMedID 3257199

  • THE STRUCTURAL GENE FOR THE M1 SUBUNIT OF RIBONUCLEOTIDE REDUCTASE MAPS TO CHROMOSOME-11, BAND P15, IN HUMAN AND TO CHROMOSOME-7 IN MOUSE EXPERIMENTAL CELL RESEARCH Brissenden, J. E., Caras, I., Thelander, L., FRANCKE, U. 1988; 174 (1): 302-308

    Abstract

    The genes for the M1 subunit of the enzyme ribonucleotide reductase have been mapped in the human and the murine species by use of two independently derived mouse cDNA clones. Southern blot analysis of rodent x human somatic cell hybrid DNAs confirmed the assignment of RRM1 to the short arm of human chromosome 11. In situ hybridization to human metaphase chromosomes revealed a peak of silver grains over the distal third of band 11p15, a region corresponding to subbands p15.4----p15.5. The mouse Rrml locus was assigned to chromosome 7, where it forms part of a conserved syntenic group of at least seven other genes assigned to human chromosome band 11p15.

    View details for Web of Science ID A1988L704600030

    View details for PubMedID 3275546

  • Molecular Analysis of the cDNA for Human SPARC/Osteonectin/BM-40: Sequence, Expression, and Localization of the Gene to Chromosome 5q31-q33 GENOMICS Swaroop, A., Hogan, B. L., Francke, U. 1988; 2 (1): 37-47
  • IDENTIFICATION OF A CDNA-ENCODING A PARATHYROID HORMONE-LIKE PEPTIDE FROM A HUMAN-TUMOR ASSOCIATED WITH HUMORAL HYPERCALCEMIA OF MALIGNANCY PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Mangin, M., Webb, A. C., Dreyer, B. E., POSILLICO, J. T., Ikeda, K., Weir, E. C., Stewart, A. F., Bander, N. H., Milstone, L., Barton, D. E., FRANCKE, U., Broadus, A. E. 1988; 85 (2): 597-601

    Abstract

    Humoral hypercalcemia of malignancy is a common paraneoplastic syndrome that appears to be mediated in many instances by a parathyroid hormone-like peptide. Poly(A)+ RNA from a human renal carcinoma associated with this syndrome was enriched by preparative electrophoresis and used to construct an enriched cDNA library in phage lambda gt10. The library was screened with a codon-preference oligonucleotide synthesized on the basis of a partial N-terminal amino acid sequence from a human tumor-derived peptide, and a 2.0-kilobase cDNA was identified. The cDNA encodes a 177 amino acid protein consisting of a 36 amino acid leader sequence and a 141 amino acid mature peptide. The first 13 amino acids of the deduced sequence of the mature peptide display strong homology to human PTH, with complete divergence thereafter. RNA blot-hybridization analysis revealed multiple transcripts in mRNA from tumors associated with the humoral syndrome and also in mRNA from normal human keratinocytes. Southern blot analysis of genomic DNA from humans and rodents revealed a simple pattern compatible with a single-copy gene. The gene has been mapped to chromosome 12.

    View details for Web of Science ID A1988L864700064

    View details for PubMedID 2829195

  • COMPARISON OF NONERYTHROID ALPHA-SPECTRIN GENES REVEALS STRICT HOMOLOGY AMONG DIVERSE SPECIES MOLECULAR AND CELLULAR BIOLOGY Leto, T. L., FORTUGNOERIKSON, D., Barton, D., YANGFENG, T. L., FRANCKE, U., Harris, A. S., Morrow, J. S., Marchesi, V. T., Benz, E. J. 1988; 8 (1): 1-9

    Abstract

    The spectrins are a family of widely distributed filamentous proteins. In association with actin, spectrins form a supporting and organizing scaffold for cell membranes. Using antibodies specific for human brain alpha-spectrin (alpha-fodrin), we have cloned a rat brain alpha-spectrin cDNA from an expression library. Several closely related human clones were also isolated by hybridization. Comparison of sequences of these and other overlapping nonerythroid and erythroid alpha-spectrin genes demonstrated that the nonerythroid genes are strictly conserved across species, while the mammalian erythroid genes have diverged rapidly. Peptide sequences deduced from these cDNAs revealed that the nonerythroid alpha-spectrin chain, like the erythroid spectrin, is composed of multiple 106-amino-acid repeating units, with the characteristic invariant tryptophan as well as other charged and hydrophobic residues in conserved locations. However, the carboxy-terminal sequence varies markedly from this internal repeat pattern and may represent a specialized functional site. The nonerythroid alpha-spectrin gene was mapped to human chromosome 9, in contrast to the erythroid alpha-spectrin gene, which has previously been assigned to a locus on chromosome 1.

    View details for Web of Science ID A1988L482800001

    View details for PubMedID 3336352

  • Molecular analysis of the cDNA for human SPARC/osteonectin/BM-40: sequence, expression, and localization of the gene to chromosome 5q31-q33. Genomics Swaroop, A., Hogan, B. L., FRANCKE, U. 1988; 2 (1): 37-47

    Abstract

    Human cDNA clones encoding the extracellular calcium-binding, acidic glycoprotein known as SPARC, osteonectin, or BM-40 were isolated from a placental cDNA library. Two polyadenylated transcripts of 2.2 and 3.0 kb were detected in human tissues and cultured cells by Northern blot analysis, and cDNAs for both transcripts were characterized. The 2133-bp sequence of the more abundant (major) transcript contains an open reading frame for 303 amino acids. The deduced polypeptide has extensive amino acid sequence identity with mouse SPARC. The larger and minor 3.0-kb cDNA has an identical coding region but utilizes a downstream polyadenylation signal. Gene localization studies have revealed a single chromosomal site at 5q31-q33 by somatic cell hybrid analysis and in situ chromosomal hybridization. Furthermore, pulsed-field gel electrophoresis of human genomic DNA cleaved with different rare-cutting restriction enzymes and hybridized with SPARC cDNA probes revealed single or double fragments of less than 50 to about 150 kb. The evidence is consistent with a single locus for SPARC in humans. The gene was found to be differentially expressed in many human tissues and in an osteogenic sarcoma, but not in other transformed cells.

    View details for PubMedID 2838412

  • Chromosomal mapping of genes for transforming growth factors beta 2 and beta 3 in man and mouse: dispersion of TGF-beta gene family. Oncogene research Barton, D. E., FOELLMER, B. E., Du, J., Tamm, J., DERYNCK, R., FRANCKE, U. 1988; 3 (4): 323-331

    Abstract

    Human cDNA probes for two new types of transforming growth factor-beta, TGF-beta 2 and TGF-beta 3, were used for mapping their cognate genes on human and mouse chromosomes by Southern blot analysis of somatic cell hybrid lines and, for the human loci, also by in situ chromosomal hybridization. For TGF-beta 2, a single site was found on the long arm of human chromosome 1, band 1q41, and on mouse chromosome 1, most likely in the known conserved syntenic region. For TGF-beta 3, the major site of hybridization, both on Southern filters and direct chromosome preparations, was at 14q24 in humans. This region is homologous in part to mouse chromosome 12, to which the murine beta 3 locus was mapped. These results indicate a wide dispersion of the TGF-beta gene family, with genes for TGF-beta 1 previously mapped by us to human chromosome 19q and mouse chromosome 7 and for inhibins alpha, beta B and beta A to human chromosomes 2q33-qter, 2cen-q13 and 7p15-p13, respectively.

    View details for PubMedID 3226728

  • THE HUMAN U1-70K SNRNP PROTEIN - CDNA CLONING, CHROMOSOMAL LOCALIZATION, EXPRESSION, ALTERNATIVE SPLICING AND RNA-BINDING NUCLEIC ACIDS RESEARCH Spritz, R. A., Strunk, K., Surowy, C. S., Hoch, S. O., Barton, D. E., FRANCKE, U. 1987; 15 (24): 10373-10391

    Abstract

    We have isolated and sequenced cDNA clones encoding the human U1-70K snRNP protein, and have mapped this locus (U1AP1) to human chromosome 19. The gene produces two size classes of RNA, a major 1.7-kb RNA and a minor 3.9-kb RNA. The 1.7-kb species appears to be the functional mRNA; the role of the 3.9-kb RNA, which extends further in the 5' direction, is unclear. The actual size of the hU1-70K protein is probably 52 kd, rather than 70 kd. The protein contains three regions similar to known nucleic acid-binding proteins, and it binds RNA in an in vitro assay. Comparison of the cDNA sequences indicates that there are multiple subclasses of mRNA that arise by alternative pre-mRNA splicing of at least four alternative exon segments. This suggests that multiple forms of the hU1-70K protein may exist, possibly with different functions in vivo.

    View details for Web of Science ID A1987L453200020

    View details for PubMedID 2447561

  • Isolation of cDNA clones coding for rat isovaleryl-CoA dehydrogenase and assignment of the gene to human chromosome 15. Genomics Kraus, J. P., Matsubara, Y., Barton, D., Yang-Feng, T. L., GLASSBERG, R., Ito, M., Ikeda, Y., Mole, J., FRANCKE, U., Tanaka, K. 1987; 1 (3): 264-269

    Abstract

    Rat liver mRNA encoding the cytoplasmic precursor of mitochondrial isovaleryl-CoA dehydrogenase was highly enriched by polysome immunopurification using a polyclonal monospecific antibody. The purified mRNA was used to prepare a plasmid cDNA library which was screened with two oligonucleotide mixtures encoding two peptides in the amino-terminal portion of mature rat isovaleryl-CoA dehydrogenase. Thirty-one overlapping cDNA clones, spanning a region of 2.1 kbp, were isolated and characterized. The cDNA sequence of a 5'-end clone, rIVD-13 (155 bp), predicts a mitochondrial leader peptide of 30 amino acid residues and the first 18 amino acids of the mature protein. These consecutive 18 residues completely matched the amino-terminal peptide determined by automated Edman degradation of the rat enzyme. The leader peptide contains six arginines, has no acidic residues, and is particularly rich in leucine, alanine, and proline residues. Southern blot analysis of DNAs from human-rodent somatic cell hybrids with an isolated rat cDNA (2 kbp) assigned the isovaleryl-CoA dehydrogenase gene to the long arm of chromosome 15, region q14----qter. The chromosomal assignment was confirmed and further refined to bands q14----q15 by in situ hybridization of the probe to human metaphase cells. This location differs from that of the gene for medium-chain acyl-CoA dehydrogenase, a closely related enzyme, which has been previously assigned to chromosome 1.

    View details for PubMedID 3446585

  • ACTIVATION OF HUMAN ALPHA-1-ANTITRYPSIN GENES IN RAT HEPATOMA X HUMAN FIBROBLAST HYBRID CELL-LINES IS CORRELATED WITH DEMETHYLATION SOMATIC CELL AND MOLECULAR GENETICS Barton, D. E., FRANCKE, U. 1987; 13 (6): 635-644

    Abstract

    Alpha-1-antitrypsin (AAT) is the major protease inhibitor in human serum and is primarily expressed in the liver. We have studied AAT expression in fusion hybrids between a rat hepatoma line and either human fetal liver fibroblasts (series XXII) or human skin fibroblasts (series XIX). While the human AAT gene was always activated in series XXII hybrids when it was present, it was only rarely activated in series XIX hybrids. RFLP analysis revealed that both parental AAT alleles in series XIX hybrids were capable of being activated. Molecular analysis of the AAT gene in expressing and nonexpressing hybrids revealed that active AAT genes were hypomethylated, while inactive AAT genes were highly methylated. However, differences in methylation patterns were confined to the 5' end of the gene, on both sides of the first exon. DNaseI sensitivity revealed no hypersensitive sites close to active or inactive AAT genes.

    View details for Web of Science ID A1987K775200006

    View details for PubMedID 2823394

  • HUMAN PROTOONCOGENE C-KIT - A NEW CELL-SURFACE RECEPTOR TYROSINE KINASE FOR AN UNIDENTIFIED LIGAND EMBO JOURNAL Yarden, Y., Kuang, W. J., YANGFENG, T., Coussens, L., MUNEMITSU, S., Dull, T. J., Chen, E., Schlessinger, J., FRANCKE, U., Ullrich, A. 1987; 6 (11): 3341-3351

    Abstract

    Structural features of v-kit, the oncogene of HZ4 feline sarcoma virus, suggested that this gene arose by transduction and truncation of cellular sequences. Complementary DNA cloning of the human proto-oncogene coding for a receptor tyrosine kinase confirmed this possibility: c-kit encodes a transmembrane glycoprotein that is structurally related to the receptor for macrophage growth factor (CSF-1) and the receptor for platelet-derived growth factor. The c-kit gene is widely expressed as a single, 5-kb transcript, and it is localized to human chromosome 4 and to mouse chromosome 5. A c-kit peptide antibody permitted the identification of a 145,000 dalton c-kit gene product that is inserted in the cellular plasma membrane and is capable of self-phosphorylation on tyrosine residues in both human glioblastoma cells and transfected mouse fibroblasts. Our results suggest that p145c-kit functions as a cell surface receptor for an as yet unidentified ligand. Furthermore, carboxy- and amino-terminal truncations that occurred during the viral transduction process are likely to have generated the transformation potential of v-kit.

    View details for Web of Science ID A1987K569000020

    View details for PubMedID 2448137

  • RESTRICTION OF HUMAN ADENOVIRUS REPLICATION IN CHINESE-HAMSTER CELL-LINES AND THEIR HYBRIDS WITH HUMAN-CELLS VIRUS RESEARCH RADNA, R. L., FOELLMER, B., Feldman, L. A., FRANCKE, U., Ozer, H. L. 1987; 8 (4): 277-299

    Abstract

    We have found that the replication of human adenovirus (Ad2) is restricted in multiple Chinese hamster cell lines including CHO and V79. The major site of restriction involves differential accumulation of late viral proteins as demonstrated by immunofluorescence assay and polyacrylamide gel electrophoresis with and without prior immunoprecipitation. Synthesis of fiber and penton base are markedly reduced, whereas others, such as the 100K polypeptide, are synthesized efficiently. This pattern of restriction is similar to that previously reported for Ad2 infection of several monkey cell lines; however, the restriction is more marked in the Chinese hamster cell lines. The restriction is most likely due to a deficient cellular function since stable cell hybrids between V79 or CHO and human cells are permissive for virus replication. By analysis of a series of hybrids with reduced numbers of human chromosomes, fiber synthesis was correlated with the presence of the short arm of human chromosome 3. More hybrids showed restoration of fiber synthesis than production of progeny virus, suggesting that more than one unlinked function is required for the latter.

    View details for Web of Science ID A1987K998000001

    View details for PubMedID 3433922

  • CLONING, SEQUENCING, AND EXPRESSION OF THE GENE CODING FOR THE HUMAN-PLATELET ALPHA-2-ADRENERGIC RECEPTOR SCIENCE Kobilka, B. K., MATSUI, H., Kobilka, T. S., YANGFENG, T. L., FRANCKE, U., Caron, M. G., Lefkowitz, R. J., Regan, J. W. 1987; 238 (4827): 650-656

    Abstract

    The gene for the human platelet alpha 2-adrenergic receptor has been cloned with oligonucleotides corresponding to the partial amino acid sequence of the purified receptor. The identity of this gene has been confirmed by the binding of alpha 2-adrenergic ligands to the cloned receptor expressed in Xenopus laevis oocytes. The deduced amino acid sequence is most similar to the recently cloned human beta 2- and beta 1-adrenergic receptors; however, similarities to the muscarinic cholinergic receptors are also evident. Two related genes have been identified by low stringency Southern blot analysis. These genes may represent additional alpha 2-adrenergic receptor subtypes.

    View details for Web of Science ID A1987K595500029

    View details for PubMedID 2823383

  • A PARTIAL DELETION OF THE MUSCULAR-DYSTROPHY GENE TRANSMITTED TWICE BY AN UNAFFECTED MALE NATURE DARRAS, B. T., FRANCKE, U. 1987; 329 (6139): 556-558

    Abstract

    A gene of unknown function located in band Xp21 on the short arm of the human X chromosome gives rise to X-linked recessive muscular dystrophy, of either Duchenne or Becker type, when mutated. The gene encodes a large muscle-specific transcript of about 14 kilobases (kb) and its genomic size extends over more than 1,800 kb. The high mutation rate (about 10(-4) per generation) is likely to result from the large target size. Submicroscopic deletions, detectable with one or more of the dozen cloned DNA probes available for regions within the gene, constitute a significant proportion of the mutations. Because no such deletions have been found in normal individuals, it is assumed that intragenic deletions are the molecular basis of the mutations. The origin of deletions can be traced in families. With sufficient data collected, it will soon be possible to answer questions about the relative frequencies of mutations in male and female gametogenesis and about the timing of mutational events in mitotic or meiotic stages of germ cell development. We have studied a four generation family containing males who have Duchenne muscular dystrophy due to deletion of the sequence recognized by intragenic probe J-Bir. The deletion was present in two of five daughters of a woman who herself did not have the deletion. Haplotype analysis on 15 members of this family using nine informative restriction fragment length polymorphism (RFLP) markers indicated that the J-Bir deletion chromosome was transmitted from the unaffected father.

    View details for Web of Science ID A1987K320600066

    View details for PubMedID 2889145

  • The myelin-associated glycoprotein gene: mapping to human chromosome 19 and mouse chromosome 7 and expression in quivering mice. Genomics Barton, D. E., ARQUINT, M., Roder, J., Dunn, R., FRANCKE, U. 1987; 1 (2): 107-112

    Abstract

    Myelin-associated glycoprotein (MAG), a membrane glycoprotein of 100 kDa, is thought to be involved in the process of myelination. A cDNA encoding the amino-terminal half of rat MAG has recently been isolated and sequenced. We have used this cDNA in Southern blot analysis of DNA from 32 somatic cell hybrids to assign the human locus for MAG to chromosome 19 and the mouse locus to chromosome 7. Since the region of mouse chromosome 7-known to contain several other genes that are homologous to genes on human chromosome 19-also carries the quivering (qv) locus, we considered the possibility that a mutation in the MAG gene could be responsible for this neurological disorder. While MAG-specific DNA restriction fragments, mRNA, and protein from qv/qv mice were apparently normal in size and abundance, we have not ruled out the possibility that qv could be caused by a point mutation in the MAG gene.

    View details for PubMedID 2447011

  • AN INTRONLESS GENE ENCODING A POTENTIAL MEMBER OF THE FAMILY OF RECEPTORS COUPLED TO GUANINE-NUCLEOTIDE REGULATORY PROTEINS NATURE Kobilka, B. K., Frielle, T., Collins, S., YANGFENG, T., Kobilka, T. S., FRANCKE, U., Lefkowitz, R. J., Caron, M. G. 1987; 329 (6134): 75-79

    Abstract

    Plasma membrane receptors for hormones, drugs, neurotransmitters and sensory stimuli are coupled to guanine nucleotide regulatory proteins. Recent cloning of the genes and/or cDNAs for several of these receptors including the visual pigment rhodopsin, the adenylate-cyclase stimulatory beta-adrenergic receptor and two subtypes of muscarinic cholinergic receptors has suggested that these are homologous proteins with several conserved structural and functional features. Whereas the rhodopsin gene consists of five exons interrupted by four introns, surprisingly the human and hamster beta-adrenergic receptor genes contain no introns in either their coding or untranslated sequences. We have cloned and sequenced a DNA fragment in the human genome which cross-hybridizes with a full-length beta 2-adrenergic receptor probe at reduced stringency. Like the beta 2-adrenergic receptor this gene appears to be intronless, containing an uninterrupted long open reading frame which encodes a putative protein with all the expected structural features of a G-protein-coupled receptor.

    View details for Web of Science ID A1987J846700060

    View details for PubMedID 3041227

  • Ribonucleotide reductase M2 subunit sequences mapped to four different chromosomal sites in humans and mice: functional locus identified by its amplification in hydroxyurea-resistant cell lines. Genomics Yang-Feng, T. L., Barton, D. E., Thelander, L., Lewis, W. H., SRINIVASAN, P. R., FRANCKE, U. 1987; 1 (1): 77-86

    Abstract

    The sites of sequences homologous to a murine cDNA for ribonucleotide reductase (RR) subunit M2 were determined on human and murine chromosomes by Southern blot analysis of interspecies somatic cell hybrid lines and by in situ hybridization. In the human genome, four chromosomal sites carrying RRM2-related sequences were identified at 1p31----p33, 1q21----q23, 2p24----p25, and Xp11----p21. In the mouse, M2 sequences were found on chromosomes 4, 7, 12, and 13 by somatic cell hybrid studies. By Southern analysis of human hydroxyurea-resistant cells that overproduce M2 because of gene amplification, we have identified the amplified restriction fragments as those that map to chromosome 2. To further confirm the site of the functional RRM2 locus, two other cDNA clones, p5-8 and S7 (coding for ornithine decarboxylase; ODC), which are coamplified with RRM2 sequences in human and rodent hydroxyurea-resistant cell lines, were mapped by Southern and in situ hybridization. Their chromosomal map positions coincided with the region of human chromosome 2 (p24----p25) that also contains one of the four RRM2-like sequences. Since this RRM2 sequence and p5-8 and ODC are most likely part of the same amplification unit, the RRM2 structural gene can be assigned to human chromosome 2p24----p25. This region is homologous to a region of mouse chromosome 12 that also carries one of numerous ODC-like sequences. In an RRM2-overproducing mouse cell line, we found amplification of the chromosome 12-specific restriction fragments. Thus, we conclude that mouse chromosome 12 carries the functional locus for RRM2.

    View details for PubMedID 3311968

  • CYTOGENETIC AND MOLECULAR GENETIC-STUDIES OF A PATIENT WITH ATYPICAL LYMPHOID HYPERPLASIA CANCER GENETICS AND CYTOGENETICS CHENEVIXTRENCH, G., Cowan, J. M., Behm, F. G., Goorha, R., BROWN, J. A., Westin, E. H., FRANCKE, U. 1987; 27 (2): 251-259

    Abstract

    We have karyotyped cells from a lymph node of a patient with atypical lymphoid hyperplasia. Among other clonal chromosomal abnormalities, a t(2;19) translocation was observed with breakpoints at 2p11.2 and 19q13. The genes for transforming growth factor alpha and beta have been mapped to 2p11-p13 and 19q13, respectively, but Southern blot analysis did not reveal any alteration in the structure of these genes. Similarly, the kappa immunoglobulin gene, which maps to 2p11-p12 was not rearranged. In addition, Southern blot analysis using immunoglobulin and T-cell receptor genes as probes, did not demonstrate any clonality of either B or T cells. We propose that this patient represents an early, polyclonal stage of atypical hyperplasia. The chromosome changes observed may have been one of the etiologic factors causing this disorder.

    View details for Web of Science ID A1987J268500007

    View details for PubMedID 3036340

  • MOSAIC TETRASOMY 12P - 4 NEW CASES, AND CONFIRMATION OF THE CHROMOSOMAL ORIGIN OF THE SUPERNUMERARY CHROMOSOME IN ONE OF THE ORIGINAL PALLISTER-MOSAIC SYNDROME CASES AMERICAN JOURNAL OF MEDICAL GENETICS WARBURTON, D., ANYANEYEBOA, K., FRANCKE, U. 1987; 27 (2): 275-283

    Abstract

    Four new cases are reported in which mosaicism for a supernumerary chromosome interpreted as an isochromosome for 12p [i(12p)] is present. In 2 cases seen in early childhood the mosaicism was present at a low level in peripheral blood and was documented in one case to be present with a higher frequency in fibroblast cultures from skin. These cases have clinical features compatible with those in previously reported cases of the Teschler-Nicola/Killian syndrome, many of whom have now been found to be mosaic for a similar i(12p) chromosome in fibroblast cultures. One case was diagnosed prenatally from amniotic fluid culture. The fourth case was a neonatal death, in which fibroblast cultures were established from muscle and increased activity of LDH-B was demonstrated, supporting the theory that the origin of the additional chromosome was from 12p. Loss of the cell line with the supernumerary chromosome occurs after long-term fibroblast culture. Previously unpublished studies showing increased LDH-B activity in case 1 of Pallister-Mosaic syndrome originally reported in 1977 are also reported. It is of interest that our 2 cases which did not survive birth and one previously published case diagnosed prenatally had diaphragmatic herniae.

    View details for Web of Science ID A1987H339800004

    View details for PubMedID 3605213

  • MOLECULAR ANALYSIS AND CHROMOSOMAL MAPPING OF AMPLIFIED GENES ISOLATED FROM A TRANSFORMED MOUSE 3T3-CELL LINE SOMATIC CELL AND MOLECULAR GENETICS CAHILLYSNYDER, L., YANGFENG, T., FRANCKE, U., George, D. L. 1987; 13 (3): 235-244

    Abstract

    We are exploring the origin and function of amplified DNA sequences associated with double minutes (DMs) in a spontaneously transformed derivative of mouse 3T3 cells. Toward that goal, we have constructed a cDNA library using RNA from these cells and have isolated cDNA clones representing sequences that are amplified and overexpressed in these 3T3-DM cells. From results of Northern- and Southern-blot analyses, we conclude that these cDNAs represent two distinct genes, which we have designated mdm-1 and mdm-2. Using DNAs from a panel of Chinese hamster-mouse somatic cell hybrids together with in situ hybridization protocols for gene mapping studies, we have found that these DM-associated, amplified DNA sequences originate from mouse chromosome 10, region C1-C3. Sequences homologous to mdm-1 and mdm-2 are present in the genomes of several species examined, including that of man.

    View details for Web of Science ID A1987H903500005

    View details for PubMedID 3474784

  • PRENATAL-DIAGNOSIS AND DETECTION OF CARRIERS WITH DNA PROBES IN DUCHENNES MUSCULAR-DYSTROPHY NEW ENGLAND JOURNAL OF MEDICINE DARRAS, B. T., HARPER, J. F., FRANCKE, U. 1987; 316 (16): 985-992

    Abstract

    We performed genetic analyses for the prenatal diagnosis of Duchenne's muscular dystrophy and detection of the carrier state in five families with seven pregnancies at risk for the disease. As genetic markers for the disorder, we used DNA-sequence polymorphisms detected with 12 different DNA probes derived from the vicinity of the Duchenne's muscular dystrophy locus or from within the gene, on the X chromosome. One male fetus of a proved carrier mother was predicted to be unaffected, and this was confirmed after birth. Another male fetus was predicted to be unaffected (probability, 95 percent or greater), although a crossover event had been identified in a region of the X chromosome thought to be distal to the Duchenne gene. Unfortunately, an elevated serum creatine kinase level after birth indicated that the infant had inherited the Duchenne mutation. Three male fetuses predicted to be affected with 66 percent or 95 percent probabilities were aborted, and the presence of the DNA-marker alleles was confirmed in fetal tissues. In one family, in which the maternal grandparents were unavailable, the initial genetic interpretation had to be revised after a second male fetus was analyzed with intragenic probes. Our experience suggests that despite the large number of intragenic and flanking DNA polymorphisms available, uncertainties often remain in the prenatal diagnosis of Duchenne's muscular dystrophy. Pitfalls are presented by the large size of the region in which Duchenne's mutations can occur. Crossover events in this region, which result in an exchange of DNA between two X chromosomes, can render DNA-marker studies inaccurate. Also, an autosomal recessive mutation can produce the same clinical picture.

    View details for Web of Science ID A1987G820000004

    View details for PubMedID 3561454

  • CONGENITAL ADRENAL HYPOPLASIA, MYOPATHY, AND GLYCEROL KINASE-DEFICIENCY - MOLECULAR GENETIC-EVIDENCE FOR DELETIONS AMERICAN JOURNAL OF HUMAN GENETICS FRANCKE, U., HARPER, J. F., DARRAS, B. T., Cowan, J. M., McCabe, E. R., Kohlschutter, A., Seltzer, W. K., Saito, F., Goto, J., Harpey, J. P., Wise, J. E. 1987; 40 (3): 212-227

    Abstract

    Glycerol kinase deficiency (GKD) is an X-linked recessive trait that occurs in association with congenital adrenal hypoplasia (AH) and developmental delay with or without congenital dystrophic myopathy. Several such patients have recently been reported to have cytological deletions of chromosome region Xp21 and/or of DNA markers that map near the locus for Duchenne muscular dystrophy (DMD) in band Xp21. We have examined the initial family reported in the literature and, using prometaphase chromosome studies and Southern blot analysis with 13 different DNA probes derived from band Xp21, have found no deletions within this region of the X chromosome. When DNA samples from six other unrelated affected males were analyzed, four of them were found to have different-size deletions within Xp21. Thus, the form of GKD associated with AH and dystrophic myopathy exhibits significant genetic heterogeneity at the DNA level. No deletions were detected in two patients with isolated GK deficiency. Comparison of our molecular studies of unrelated patients with deletions of DNA segments allows us to define the region of Xp21 (between probes J-Bir and L1.4) that most likely contains the genes for GKD and AH. This location is distal to the DMD locus. The patients with progressive muscular dystrophy tended to have larger deletions that include markers known to derive from the DMD locus, while GKD/AH/dystrophic-myopathy patients without current evidence of deletion seemed to have a milder, nonprogressive form of congenital myopathy.

    View details for Web of Science ID A1987H085400002

    View details for PubMedID 2883886

  • MOLECULAR ANALYSIS OF SYNAPSIN-I, A CANDIDATE GENE FOR RETT SYNDROME BRAIN & DEVELOPMENT DeGennaro, L. J., MCCAFFERY, C. A., Kirchgessner, C. U., YANGFENG, T. L., FRANCKE, U. 1987; 9 (5): 469-474

    Abstract

    The characteristics of Rett syndrome suggest that it is an X-linked neurodegenerative disorder. Laboratory investigations to date have not revealed any metabolic abnormalities in affected individuals. Synapsin I is a neuron-specific protein thought to play a fundamental role in neuronal function. In this report we summarize the circumstantial evidence suggesting that a defect in synapsin I gene structure or expression might be involved in Rett syndrome. This evidence includes analysis of structural and functional aspects of synapsin I primary structure, characterization of synapsin I messenger RNAs, location of the synapsin I gene on the human X chromosome and preliminary analysis of synapsin I gene structure in Rett individuals.

    View details for Web of Science ID A1987L502800006

    View details for PubMedID 3124657

  • THE STRUCTURAL GENE OF HUMAN DNA POLYMERASE-ALPHA (POLA) IS LOCATED ON THE SHORT ARM OF THE X-CHROMOSOME Wong, S. W., Wahl, A. F., Pearson, B. E., Mohandas, T., Shapiro, L. J., FRANCKE, U., Korn, D., Wang, T. S. KARGER. 1987: 719–19
  • THE PHYSICAL MAP OF MUS-MUSCULUS CHROMOSOME-11 REVEALS EVOLUTIONARY RELATIONSHIPS WITH DIFFERENT SYNTENIC GROUPS OF GENES IN HOMO-SAPIENS JOURNAL OF MOLECULAR EVOLUTION MUNKE, M., FRANCKE, U. 1987; 25 (2): 134-140

    Abstract

    The physical localization of sequences homologous to three cloned genes was determined by in situ hybridization to metaphase chromosomes. Previous work had assigned the skeletal myosin heavy chain gene cluster (Myh), the functional locus for the cellular tumor antigen p53 (Trp53-1), and the cellular homologue of the viral erb-B oncogene (Erbb) to Mus musculus chromosome 11 (MMU11). Our results provide regional assignments of Myh and Trp53-1 to chromosome bands B2----C, and of Erbb to bands A1----A4. Taken together with in situ mapping of three other loci on MMU 11 (Hox-2 homeobox-containing gene cluster, the Sparc protein, and the Colla-1 collagen gene), which have been reported elsewhere, these data allowed us to construct a physical map of MMU11 and to compare it with the linkage map of this chromosome. The map positions of the homologous genes on human chromosomes suggest evolutionary relationships of distinct regions of MMU11 with six different human chromosome arms: 1p, 5q, 7p, 16p, 17p, and 17q. The delineation of conserved chromosome regions has important implications for the understanding of karyotype evolution in mammalian species and for the development of animal models of human genetic diseases.

    View details for Web of Science ID A1987H880100006

    View details for PubMedID 3116273

  • CDNA FOR THE HUMAN BETA-2-ADRENERGIC RECEPTOR - A PROTEIN WITH MULTIPLE MEMBRANE-SPANNING DOMAINS AND ENCODED BY A GENE WHOSE CHROMOSOMAL LOCATION IS SHARED WITH THAT OF THE RECEPTOR FOR PLATELET-DERIVED GROWTH-FACTOR PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Kobilka, B. K., Dixon, R. A., Frielle, T., Dohlman, H. G., BOLANOWSKI, M. A., SIGAL, I. S., YANGFENG, T. L., FRANCKE, U., Caron, M. G., Lefkowitz, R. J. 1987; 84 (1): 46-50

    Abstract

    We have isolated and sequenced a cDNA encoding the human beta 2-adrenergic receptor. The deduced amino acid sequence (413 residues) is that of a protein containing seven clusters of hydrophobic amino acids suggestive of membrane-spanning domains. While the protein is 87% identical overall with the previously cloned hamster beta 2-adrenergic receptor, the most highly conserved regions are the putative transmembrane helices (95% identical) and cytoplasmic loops (93% identical), suggesting that these regions of the molecule harbor important functional domains. Several of the transmembrane helices also share lesser degrees of identity with comparable regions of select members of the opsin family of visual pigments. We have localized the gene for the beta 2-adrenergic receptor to q31-q32 on chromosome 5. This is the same position recently determined for the gene encoding the receptor for platelet-derived growth factor and is adjacent to that for the FMS protooncogene, which encodes the receptor for the macrophage colony-stimulating factor.

    View details for Web of Science ID A1987F667900010

    View details for PubMedID 3025863

  • LOCALIZATION OF THE LDHA GENE TO 11P14-]11P15 BY INSITU HYBRIDIZATION OF AN LDHA CDNA PROBE TO 2 TRANSLOCATIONS WITH BREAKPOINTS IN 11P13 HUMAN GENETICS YANGFENG, T. L., Bruns, G. A., Carroll, A. J., Simola, K. O., FRANCKE, U. 1986; 74 (4): 331-334

    Abstract

    Lymphoblastoid cell lines established from two individuals with apparently balanced translocations involving 11p13 were used for LDHA regional localization. The karyotypes were 46,XY,t(4;11)(q21;p13) and 46,XY,t(1;11)(p22;p13). In situ hybridization of a human LDHA cDNA probe to chromosome preparations from these cell lines resulted in specific labeling over bands p14----p15 of the normal chromosomes 11 and over bands 11p14----11p15 of the derivative chromosomes 4 and 1. These results exclude LDHA from any region proximal to 11p13 and localize the gene to 11p14----11p15.

    View details for Web of Science ID A1986F310100001

    View details for PubMedID 3793094

  • GENETIC AND CYTOGENETIC LOCALIZATION OF THE HOMEO BOX CONTAINING GENES ON MOUSE CHROMOSOME-6 AND HUMAN CHROMOSOME-7 EMBO JOURNAL Bucan, M., YANGFENG, T., COLBERGPOLEY, A. M., Wolgemuth, D. J., Guenet, J. L., FRANCKE, U., Lehrach, H. 1986; 5 (11): 2899-2905

    Abstract

    Probes from the m6 homeo box cluster were mapped to mouse chromosome 6 by somatic cell genetics, in situ hybridisation, and by a Mus spretus--Mus musculus backcross mapping system. In addition, the testis-specific homeo box containing cDNA, clone, HBT-1, has been mapped using the same back-cross system and the B X D recombinant inbred strain set. Close genetic and physical linkage between the m6 cluster and HBT-1 was demonstrated, positioning these sequences to the same local cluster of homeo box containing genes. The map location of this cluster between IgK and Tcrb coincides with the morphological mutation hypodactyly (Hd). Synteny has been observed between a region of mouse chromosome 6 and the long arm of human chromosome 7 encompassing the markers Cpa, Tcrb and Try-1. Here we localise human sequences hybridising to the mouse m6 probes to the short arm of chromosome 7, breaking the region of synteny.

    View details for Web of Science ID A1986E782500023

    View details for PubMedID 2878803

  • ISOLATION OF A RANDOM COSMID CLONE, CX5, WHICH DEFINES A NEW POLYMORPHIC LOCUS DXS148 NEAR THE LOCUS FOR DUCHENNE MUSCULAR-DYSTROPHY HUMAN GENETICS Hofker, M. H., Bergen, A. A., SKRAASTAD, M. I., Bakker, E., FRANCKE, U., Wieringa, B., Bartley, J., VANOMMEN, G. J., Pearson, P. L. 1986; 74 (3): 275-279

    Abstract

    We have isolated a random cosmid cX5 (DXS148), which maps into a small Xp21 deletion associated with Duchenne muscular dystrophy (DMD), chronic granulomatous disease (CGD), retinitis pigmentosa (RP) and McLeod syndrome, cX5 maps proximally outside several other deletions associated with DMD, glycerol kinase deficiency (GK) and adrenal hypoplasia (AHC). The following order of loci is proposed: centromere-OTC-cX5 (DXS148)-754 (DXS84)-PERT87 (DXS164)/DMD-telomere. A subclone cX5.7, isolated from this cosmid, identifies an MspI RFLP, with a minor allele frequency of 35%. This probe forms an important adjunct to the existing RFLPs for family studies in Duchenne muscular dystrophy.

    View details for Web of Science ID A1986E884900014

    View details for PubMedID 2877936

  • GENES FOR SYNAPSIN-I, A NEURONAL PHOSPHOPROTEIN, MAP TO CONSERVED REGIONS OF HUMAN AND MURINE X-CHROMOSOMES PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA YANGFENG, T. L., DeGennaro, L. J., FRANCKE, U. 1986; 83 (22): 8679-8683

    Abstract

    Synapsin I is a neuron-specific phosphoprotein associated with the membranes of small synaptic vesicles. Its function is not entirely clear, but evidence points to a possible role in the regulation of neurotransmitter release. Its biosynthesis is under developmental control. Assignment of the human synapsin I gene to the X chromosome at band Xp11 was accomplished by in situ hybridization, using a rat cDNA probe. Southern blot analysis of DNAs from a panel of human-Chinese hamster somatic cell hybrids with defined regions of the human X chromosome confirmed the in situ mapping data. The mouse synapsin I gene was assigned to the X chromosome, proximal to band XD, by Southern blot analysis of Chinese hamster-mouse somatic cell hybrids with normal or rearranged mouse X chromosomes. In situ chromosomal hybridization experiments localized the mouse synapsin I gene more precisely to bands XA1----A4. These results add to the comparative gene map of mammalian species and support certain hypotheses regarding the evolutionary relationship between human and mouse X chromosomes. We hypothesize that the synapsin I gene could be mutated in human X-linked disorders with primary neuronal degeneration, such as the Rett syndrome.

    View details for Web of Science ID A1986E906700051

    View details for PubMedID 3095840

  • CHROMATID REPULSION ASSOCIATED WITH ROBERTS/SC PHOCOMELIA SYNDROME IS REDUCED IN MALIGNANT-CELLS AND NOT EXPRESSED IN INTERSPECIES SOMATIC-CELL HYBRIDS AMERICAN JOURNAL OF HUMAN GENETICS KRASSIKOFF, N. E., Cowan, J. M., Parry, D. M., FRANCKE, U. 1986; 39 (5): 618-630

    Abstract

    Different cell types from a female patient with Roberts/SC phocomelia syndrome were evaluated quantitatively for the presence of repulsion of heterochromatin and satellite regions of mitotic chromosomes. Whereas EBV-transformed lymphoblasts from an established cell line revealed these phenomena at frequencies equal to those in PHA-stimulated lymphocytes and cultured skin fibroblasts, aneuploid cells from a metastatic melanoma displayed them at 50% lower frequency. Cocultivation of the patient's fibroblasts with either an immortal Chinese hamster cell line or with a human male fibroblast strain carrying a t(4;6)(p14;q21) translocation showed that the phenomenon was not corrected or induced by a diffusible factor or by cell-to-cell contact. In each experiment, only the patient's metaphase spreads revealed chromatid repulsion. In fusion hybrids between the patient's fibroblasts and an established Chinese hamster cell line, the human chromosomes behaved perfectly normally, suggesting that the gene product which is missing or mutant in Roberts/SC phocomelia syndrome is supplied by the Chinese hamster genome.

    View details for Web of Science ID A1986F303200007

    View details for PubMedID 3788975

  • A VARIABLE TANDEM REPEAT LOCUS MAPPED TO CHROMOSOME BAND 10Q26 IS AMPLIFIED AND REARRANGED IN LEUKOCYTE DNAS OF 2 CANCER-PATIENTS NUCLEIC ACIDS RESEARCH COLB, M., YANGFENG, T., FRANCKE, U., MERMER, B., PARKINSON, D. R., Krontiris, T. G. 1986; 14 (20): 7929-7937

    Abstract

    A highly polymorphic locus associated with the variable tandem repetition of a 35 bp consensus sequence was mapped to chromosome 10, band q26. Examination of leukocyte DNA from a cancer patient revealed the twenty-fold amplification of one allelic fragment of this locus, while the other allelic fragment demonstrated a normal copy number. In another patient, Southern blotting of leukocyte DNA detected the deletion of the 3'-flanking region from one tandem repeat allele. These results indicate that variable tandem repeats may mark highly unstable regions of DNA in the human genome which can be altered by changes more extensive than simple tandem repeat variation.

    View details for Web of Science ID A1986E640600009

    View details for PubMedID 3022240

  • MULTIPLE CHROMOSOMAL REARRANGEMENTS IN A SPONTANEOUSLY ARISING T(6-7) RAT IMMUNOCYTOMA JUXTAPOSE C-MYC AND IMMUNOGLOBULIN HEAVY-CHAIN SEQUENCES PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Pear, W. S., Ingvarsson, S., Steffen, D., MUNKE, M., FRANCKE, U., Bazin, H., Klein, G., SUMEGI, J. 1986; 83 (19): 7376-7380

    Abstract

    Spontaneously arising immunocytomas in Lou/Wsl rats contain a consistent translocation between chromosomes 6 and 7. The c-myc gene has been localized to chromosome 7 and has been shown to be rearranged in the majority of the rat immunocytomas. We now report the cloning of the rearranged 11-kilobase EcoRI c-myc fragment from the IgE-secreting IR75 tumor. Sequence analysis revealed that the cytogenetically visible t(6;7) translocation must have involved several events in this tumor. One event has led to the juxtaposition of c-myc and the switch mu region, in a head-to-head orientation. The breakpoint is approximately 850 base pairs upstream from the proximal c-myc promoter on chromosome 7. This area is distinct from the more common mouse plasmacytoma- and Burkitt lymphoma-associated translocation breakpoints and also differs from the known murine retroviral insertion sites. A second rearrangement has led to the transposition of sequences upstream from the switch gamma 1 region to the c-myc-distant end of the switch mu region, tail-to-tail. This requires at least two events, including one inversion. In addition to showing that identical loci (c-myc, immunoglobulin) are juxtaposed via chromosomal translocations in three different tumors (Burkitt lymphoma, mouse plasmacytoma, and rat immunocytoma) in different species (human, mouse, and rat), the multiple rearrangements in IR75 and some other tumors emphasize the selective value of c-myc activation by an immunoglobulin locus in the tumorigenic process.

    View details for Web of Science ID A1986E302500053

    View details for PubMedID 3020544

  • INSULIN-LIKE GROWTH FACTOR-I RECEPTOR PRIMARY STRUCTURE - COMPARISON WITH INSULIN-RECEPTOR SUGGESTS STRUCTURAL DETERMINANTS THAT DEFINE FUNCTIONAL SPECIFICITY EMBO JOURNAL Ullrich, A., Gray, A., Tam, A. W., YANGFENG, T., Tsubokawa, M., Collins, C., HENZEL, W., LeBon, T., Kathuria, S., Chen, E., Jacobs, S., FRANCKE, U., RAMACHANDRAN, J., FUJITAYAMAGUCHI, Y. 1986; 5 (10): 2503-2512

    Abstract

    To identify structural characteristics of the closely related cell surface receptors for insulin and IGF-I that define their distinct physiological roles, we determined the complete primary structure of the human IGF-I receptor from cloned cDNA. The deduced sequence predicts a 1367 amino acid receptor precursor, including a 30-residue signal peptide, which is removed during translocation of the nascent polypeptide chain. The 1337 residue, unmodified proreceptor polypeptide has a predicted Mr of 151,869, which compares with the 180,000 Mr IGF-I receptor precursor. In analogy with the 152,784 Mr insulin receptor precursor, cleavage of the Arg-Lys-Arg-Arg sequence at position 707 of the IGF-I receptor precursor will generate alpha (80,423 Mr) and beta (70,866 Mr) subunits, which compare with approximately 135,000 Mr (alpha) and 90,000 Mr (beta) fully glycosylated subunits.

    View details for Web of Science ID A1986E419000013

    View details for PubMedID 2877871

  • STRUCTURE OF THE RECEPTOR FOR PLATELET-DERIVED GROWTH-FACTOR HELPS DEFINE A FAMILY OF CLOSELY RELATED GROWTH-FACTOR RECEPTORS NATURE Yarden, Y., Escobedo, J. A., Kuang, W. J., YANGFENG, T. L., Daniel, T. O., TREMBLE, P. M., CHEN, E. Y., ANDO, M. E., HARKINS, R. N., FRANCKE, U., Fried, V. A., Ullrich, A., Williams, L. T. 1986; 323 (6085): 226-232

    Abstract

    The primary structure of the receptor for platelet-derived growth factor (PDGF), determined by means of cloning a cDNA that encodes the murine pre-PDGF receptor, is closely related to that of the v-kit oncogene product and the receptor for macrophage colony stimulating factor (CSF-1). Common structural features include the presence of long sequences that interrupt the tyrosine-specific protein kinase domains of each molecule. The PDGF and CSF-1 receptors also share a characteristic distribution of extracellular cysteine residues. Ubiquitin is covalently bound to the purified PDGF receptor, the human gene for which is on chromosome 5.

    View details for Web of Science ID A1986E040800051

    View details for PubMedID 3020426

  • MOLECULAR-CLONING OF CDNAS ENCODING RAT AND HUMAN MEDIUM-CHAIN ACYL-COA DEHYDROGENASE AND ASSIGNMENT OF THE GENE TO HUMAN CHROMOSOME-1 PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Matsubara, Y., Kraus, J. P., YANGFENG, T. L., FRANCKE, U., Rosenberg, L. E., Tanaka, K. 1986; 83 (17): 6543-6547

    Abstract

    Rat liver mRNA encoding the precursor of medium-chain acyl-CoA dehydrogenase was purified to near homogeneity by polysome immunoadsorption using a polyclonal, monospecific antibody. A single-stranded, 32P-labeled cDNA probe was synthesized using the enriched mRNA as template and was used to screen directly 15,000 colonies from a total rat liver cDNA library constructed in pBR322. One clone [600 base pairs (bp)] was positively identified by hybrid-selected translation combined with mitochondrial processing of translated products. Using the isolated rat cDNA as probe, 43,000 colonies from a human liver cDNA library were screened. Three overlapping clones (1100 bp, 500 bp, and 400 bp) were isolated and positively identified by hybrid-selected translation. The largest human cDNA clone was subcloned into the transcription vector pGEM-2, which contains a bacteriophage T7 RNA polymerase promoter. In vitro transcription of this recombinant, followed by in vitro translation, showed that the cDNA clone coded for approximately 80% of the medium-chain acyl-CoA dehydrogenase protein. The sizes of rat and human mRNAs encoding the precursor of medium-chain acyl-CoA dehydrogenase were 2.2 and 2.4 kilobases long, respectively, as determined by blot hybridization analysis of electrophoretically fractionated poly(A)+ RNA. Southern blot analysis of DNAs from human-rodent somatic cell hybrids with an isolated human cDNA assigned the gene coding for this enzyme to the short arm of chromosome 1, band p31. The chromosomal assignment was confirmed by in situ hybridization of the probe to human metaphase cells. Direct screening of cDNA libraries using a highly enriched mRNA to generate a probe, as demonstrated in this study, may provide the most rapid and convenient approach to cDNA cloning of low-abundance mRNAs.

    View details for Web of Science ID A1986D837900065

    View details for PubMedID 3462713

  • SEQUENTIAL EXPRESSION OF MURINE HOMEO BOX GENES DURING F9-EC CELL-DIFFERENTIATION EMBO JOURNAL Breier, G., Bucan, M., FRANCKE, U., COLBERGPOLEY, A. M., GRUSS, P. 1986; 5 (9): 2209-2215

    Abstract

    We have isolated and characterized a previously unknown member of the murine homeo box family. The new locus, m31, is located on chromosome 15 and is more homologous to sequences contained in the Drosophila homeotic gene, Antp, than to any other known murine homeo box. We show that this gene encodes a 2.7 kb mRNA which is expressed during mouse embryogenesis and during differentiation of F9 teratocarcinoma cells into parietal endoderm cells. The transcript appears late in this differentiation process and is most abundant at a time when the F9 cells begin to express tissue-specific markers and the expression of another murine homeo box gene, m6, has decreased.

    View details for Web of Science ID A1986E180400023

    View details for PubMedID 2877873

  • MULTIPLE, DISTINCT FORMS OF BOVINE AND HUMAN PROTEIN-KINASE-C SUGGEST DIVERSITY IN CELLULAR SIGNALING PATHWAYS SCIENCE Coussens, L., Parker, P. J., Rhee, L., YANGFENG, T. L., Chen, E., Waterfield, M. D., FRANCKE, U., Ullrich, A. 1986; 233 (4766): 859-866

    Abstract

    A new family of protein kinase C-related genes has been identified in bovine, human, and rat genomes. The alpha-, beta-, and gamma-type protein kinase sequences are highly homologous, include a kinase domain, and potential calcium-binding sites, and they contain interspersed variable regions. The corresponding genes are located on distinct human chromosomes; the possibility of even greater genetic complexity of this gene family is suggested by Northern and Southern hybridization analyses.

    View details for Web of Science ID A1986D609100019

    View details for PubMedID 3755548

  • DEVELOPMENTAL AND TRANSFORMATION-SENSITIVE EXPRESSION OF THE SPARC GENE ON MOUSE CHROMOSOME-11 EMBO JOURNAL Mason, I. J., Murphy, D., MUNKE, M., FRANCKE, U., Elliott, R. W., HOGAN, B. L. 1986; 5 (8): 1831-1837

    Abstract

    SPARC is a Mr 43,000 secreted, acidic, cysteine-rich glycoprotein homologous to 43K bovine endothelial 'culture shock' protein. We show here that it is encoded by a single gene localized to the central region of mouse chromosome 11. During development SPARC mRNA is expressed at higher levels in all the extra-embryonic tissues than in the fetus. Highest levels are found in the parietal endoderm, while visceral endoderm has approximately 6-fold less. This differential expression is also seen in F9 teratocarcinoma cells treated with retinoic acid under conditions in which they give rise to either parietal or visceral endoderm. The 20-fold increase seen during differentiation into parietal endoderm is due, at least in part, to an increase in gene transcription. We also report SPARC expression in a variety of adult tissues and cultured cells, and present evidence that a decrease accompanies the transformation of fibroblast cell lines.

    View details for Web of Science ID A1986D534000013

    View details for PubMedID 3758028

  • ANALYSIS OF DELETIONS IN DNA FROM PATIENTS WITH BECKER AND DUCHENNE MUSCULAR-DYSTROPHY NATURE KUNKEL, L. M., HEJTMANCIK, J. F., CASKEY, C. T., SPEER, A., Monaco, A. P., MIDDLESWORTH, W., COLLETTI, C. A., BERTELSON, C., Muller, U., BRESAN, M., SHAPIRO, F., TANTRAVAHI, U., SPEER, J., LATT, S. A., Bartlett, R., Pericak-Vance, M. A., Roses, A. D., THOMPSON, W. V., Ray, P. N., WORTON, R. G., FISCHBECK, K. H., GALLANO, P., COULON, M., DUROS, C., BOUE, J., JUNIEN, C., Chelly, J., HAMARD, G., JEANPIERRE, M., Lambert, M., KAPLAN, J. C., EMERY, A., DORKINS, H., McGLADE, S., Davies, K. E., BOEHM, C., ARVEILER, B., LEMAIRE, C., MORGAN, G. J., DENTON, M. J., AMOS, J., BOBROW, M., BENHAM, F., BOSWINKEL, E., COLE, C., DUBOWITZ, V., Hart, K., HODGSON, S., Johnson, L., Walker, A., Roncuzzi, L., FERLINI, A., NOBILE, C., Romeo, G., WILCOX, D. E., AFFARA, N. A., FERGUSON-SMITH, M. A., LINDOLF, M., Kaariainen, H., de la Chapelle, A., IONASESCU, V., SEARBY, C., IONASESCU, R., Bakker, E., von Ommen, G. J., Pearson, P. L., GREENBERG, C. R., HAMERTON, J. L., WROGEMANN, K., DOHERTY, R. A., POLAKOWSKA, R., HYSER, C., QUIRK, S., Thomas, N., HARPER, J. F., DARRAS, B. T., FRANCKE, U. 1986; 322 (6074): 73-77

    Abstract

    Duchenne muscular dystrophy (DMD) is an X-linked recessive genetic disorder for which the biochemical defect is as yet unknown. Recently, two cloned segments of human X-chromosome DNA have been described which detect structural alterations within or near the genetic locus responsible for the disorder. Both of these cloned segments were described as tightly linked to the locus and were capable of detecting deletions in the DNA of boys affected with DMD. In an attempt to determine more precisely the occurrence of these deletions within a large population of DMD patients and the accuracy of one of the segments, DXS164 (pERT87), in determining the inheritance of the DMD X chromosome, the subclones 1, 8 and 15 were made available to many investigators throughout the world. Here we describe the combined results of more than 20 research laboratories with respect to the occurrence of deletions at the DXS164 locus in DNA samples isolated from patients with DMD and Becker muscular dystrophy (BMD). The results indicate that the DXS164 locus apparently recombines with DMD 5% of the time, but is probably located between independent sites of mutation which yield DMD. The breakpoints of some deletions are delineated within the DXS164 locus, and it is evident that the deletions at the DMD locus are frequent and extremely large.

    View details for Web of Science ID A1986D077300064

    View details for PubMedID 3014348

  • HUMAN TISSUE-TYPE PLASMINOGEN-ACTIVATOR GENE LOCATED NEAR CHROMOSOMAL BREAKPOINT IN MYELOPROLIFERATIVE DISORDER AMERICAN JOURNAL OF HUMAN GENETICS YANGFENG, T. L., Opdenakker, G., Volckaert, G., FRANCKE, U. 1986; 39 (1): 79-87

    Abstract

    Plasminogen activators (PA) convert the inactive proenzyme plasminogen into plasmin, which is involved in the process of fibrinolysis, tissue remodeling, and cell migration. There are two distinct forms of PA: urokinase (u-PA) and tissue-type plasminogen activator (t-PA). t-PA has higher affinity for fibrin and is the main form involved in thrombolysis. By in situ chromosomal hybridization and Southern blot analysis of somatic cell hybrid DNA, we have assigned the human t-PA gene to chromosome 8, bands 8p12----q11.2. We have detected a common EcoRI restriction fragment length polymorphism within the t-PA gene that thus provides a precisely localized highly informative marker for genetic linkage studies. The t-PA gene localization coincides with a translocation breakpoint observed in myeloproliferative disorders. Whereas leukemic cells usually secrete both types of PA, a correlation exists between acute myeloid leukemic cells that release only t-PA and failure to respond to chemotherapy.

    View details for Web of Science ID A1986D961500007

    View details for PubMedID 3092643

  • ELUSIVE CHROMOSOME-ANOMALIES HOSPITAL PRACTICE FRANCKE, U. 1986; 21 (5): 175-?

    View details for Web of Science ID A1986C340700015

    View details for PubMedID 3084511

  • TRANSFORMING GROWTH-FACTOR BETA-GENE MAPS TO HUMAN-CHROMOSOME 19 LONG ARM AND TO MOUSE CHROMOSOME-7 SOMATIC CELL AND MOLECULAR GENETICS Fujii, D., Brissenden, J. E., DERYNCK, R., FRANCKE, U. 1986; 12 (3): 281-288

    Abstract

    Transforming growth factors (TGF) are defined as biologically active polypeptides which reversibly confer the transformed phenotype onto untransformed cultured cells. They have been subdivided into two classes: type alpha and type beta TGFs. TGF-beta acts synergistically with TGF-alpha in inducing phenotypic transformation. TGF-beta can also act as a negative autocrine growth factor. A human 1050-bp EcoRi cDNA fragment was used to map the human locus for TGF-beta by Southern blotting of DNA prepared from 17 human X Chinese hamster somatic cell hybrids. The human-specific restriction fragments segregated with human chromosome 19 in all of 14 informative hybrids. All other human chromosomes were discordant with the TGF-beta bands in at least four hybrids. After in situ hybridization of the tritiated TGF-beta probe to normal human metaphase spreads, 151 silver grains were scored in 54 cells. Of 24 grains over chromosome 19, 16 grains (11%) lay over region 19q13.1----q13.3. Of the 54 cells analyzed, 16 (30%) had label over region 19q13.1----q13.3. Thus, TGFB is assigned to chromosome 19, subbands q13.1----q13.3. The Tgf-beta locus in the mouse was mapped to chromosome 7 by hybridizing a murine cDNA probe to a Chinese hamster X mouse hybrid panel. Human chromosome 19 and proximal mouse chromosome 7 share another four homologous loci.

    View details for Web of Science ID A1986C737700008

    View details for PubMedID 3459257

  • AN EXPRESSED BETA-TUBULIN GENE, TUBB, IS LOCATED ON THE SHORT ARM OF HUMAN CHROMOSOME-6 AND 2 RELATED SEQUENCES ARE DISPERSED ON CHROMOSOME-8 AND CHROMOSOME-13 EXPERIMENTAL CELL RESEARCH FLOYDSMITH, G., DEMARTINVILLE, B., FRANCKE, U. 1986; 163 (2): 539-548

    Abstract

    The chromosomal assignments of an expressed beta-tubulin gene and two related sequences have been determined by Southern blot analysis of DNA from a panel of human x Chinese hamster somatic cell hybrids cleaved with Hind III or EcoRI. Probes containing the 3' untranslated regions of the expressed gene M40 and of pseudogene 21 beta were used to localize the M40 sequence (gene symbol TUBB) to chromosome 6 region 6p21----6pter, the 21 beta pseudogene (TUBBP1) to chromosome 8 region 8q21----8pter and a third related sequence (TUBBP2) to chromosome 13. Asynteny of expressed genes and related processed pseudogenes has now been demonstrated for several gene families.

    View details for Web of Science ID A1986A705700026

    View details for PubMedID 3007184

  • CLONING AND SCREENING WITH NANOGRAM AMOUNTS OF IMMUNOPURIFIED MESSENGER-RNAS - CDNA CLONING AND CHROMOSOMAL MAPPING OF CYSTATHIONINE BETA-SYNTHASE AND THE BETA-SUBUNIT OF PROPIONYL-COA CARBOXYLASE PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Kraus, J. P., Williamson, C. L., Firgaira, F. A., YANGFENG, T. L., MUNKE, M., FRANCKE, U., Rosenberg, L. E. 1986; 83 (7): 2047-2051

    Abstract

    We have developed conditions for efficient cDNA cloning of nanogram amounts of purified mRNAs coding for cystathionine beta-synthase [L-serine hydro-lyase (adding homocysteine), EC 4.2.1.22] and for the cytosolic precursors of mitochondrial ornithine transcarbamylase (carbamoylphosphate:L-ornithine carbamoyltransferase, EC 2.1.3.3) and the beta subunit of propionyl-CoA carboxylase [propanoyl-CoA: carbon-dioxide ligase (ADP-forming), EC 6.4.1.3]. The three mRNAs, prepared by sequential immunoselection from the same batch of rat liver polysomes, were pooled (20 ng each), and cDNA was synthesized by using avian reverse transcriptase. The second DNA strand was prepared by "nick-translation repair" of the cDNA . mRNA hybrid with RNase H, polymerase I, and DNA ligase from Escherichia coli. The double-stranded (ds) DNA was tailed with deoxycytidine residues, annealed with Pst I-cut/dG-tailed pBR322, and used to transform E. coli. The library generated by this three-step procedure contained 5000 independent colonies. A 550-base-pair (bp) cDNA clone of the beta subunit of propionyl-CoA carboxylase was detected by hybrid-selected translation; it was then used to screen the library for longer cDNAs. Two hybridizing cDNAs, 1200 and 1000 bp long with a 200-bp overlap, representing together a full-length copy of the coding region and 446 bp of 3' untranslated sequence, were recovered. Each plasmid mapped to the region q13.3----q22 of human chromosome 3. Cystathionine beta-synthase clones were obtained by screening the library with a single-stranded [32P]cDNA prepared directly from the highly purified synthase mRNA by reverse transcriptase. The longest hybridizing cDNA of 1700 bp was used in hybrid-selected translation and detected a polypeptide of 63 kDa, identical in size to rat liver synthase. In situ hybridization of this cDNA to q22 of human chromosome 21 confirmed two previous tentative assignments of the synthase locus to this chromosome.

    View details for Web of Science ID A1986A844300018

    View details for PubMedID 3457373

  • THE HUMAN TYROSINE AMINOTRANSFERASE GENE MAPPED TO THE LONG ARM OF CHROMOSOME-16 (REGION 16Q22-]Q24) BY SOMATIC-CELL HYBRID ANALYSIS AND INSITU HYBRIDIZATION HUMAN GENETICS Barton, D. E., YANGFENG, T. L., FRANCKE, U. 1986; 72 (3): 221-224

    Abstract

    Tyrosine aminotransferase (TAT; EC 2.6.1.5) is a liver enzyme involved in amino acid metabolism and gluconeogenesis. Low levels of TAT have been implicated in several inherited disorders, particularly tyrosinemia II (Richner-Hanhart syndrome). We have determined the chromosomal location of the human TAT gene by Southern blot hybridization analysis of DNAs from 18 human X rodent hybrid cell lines, using a rat cDNA probe. The results indicate that the TAT gene maps to chromosome 16. Analysis of two hybrids containing a rearranged chromosome 16 allowed assignment of the TAT locus to 16q22----24. In situ hybridization to human metaphase chromosomes confirmed this regional assignment.

    View details for Web of Science ID A1986A661400006

    View details for PubMedID 2870017

  • THE INVOLVEMENT OF 6P IN MELANOMA CANCER GENETICS AND CYTOGENETICS Cowan, J. M., Halaban, R., Lane, A. T., FRANCKE, U. 1986; 20 (3-4): 255-261

    Abstract

    Karyotype analysis of first passage cells from a melanoma, occurring in a patient with a giant congenital nevus, revealed monosomy of chromosomes #6 and #11, and trisomy of chromosomes #8 and #22. Five marker chromosomes were present, including two ring chromosomes. The origin of three of the marker chromosomes was determined, and all were found to contain the region 6p21.1-6p23. The ring chromosomes were also thought to contain this region, to which the major histocompatibility locus has been mapped. The significance of these findings is discussed in the light of recent reports of chromosome studies of melanoma cells.

    View details for Web of Science ID A1986AZK1300008

    View details for PubMedID 3943065

  • THE TERMINAL DEOXYNUCLEOTIDYLTRANSFERASE GENE IS LOCATED ON HUMAN CHROMOSOME-10 (10Q23-]Q24) AND ON MOUSE CHROMOSOME-19 CYTOGENETICS AND CELL GENETICS YANGFENG, T. L., Landau, N. R., Baltimore, D., FRANCKE, U. 1986; 43 (3-4): 121-126

    Abstract

    Terminal deoxynucleotidyltransferase (TdT) is a DNA polymerase expressed in immature lymphocytes of the thymus and bone marrow, as well as certain leukemic cells. Chromosomal assignment of the gene coding for human TdT was accomplished by in situ hybridization of a 3H-labeled cDNA probe to human chromosome preparations and by Southern blot analysis of somatic cell hybrid DNAs. The human TdT gene was mapped to the region q23----q24 of chromosome 10. Breaks at this site have been reported in different translocations in human leukemias. The mouse TdT gene was assigned to chromosome 19 by Southern blot analysis of mouse X Chinese hamster somatic cell hybrids. This result adds a fourth locus to the conserved syntenic group on mouse chromosome 19 and human chromosome 10.

    View details for Web of Science ID A1986G020900001

    View details for PubMedID 3467897

  • THE MURINE HOX-2 CLUSTER OF HOMEO BOX CONTAINING GENES MAPS DISTAL ON CHROMOSOME-11 NEAR THE TAIL-SHORT (TS) LOCUS CYTOGENETICS AND CELL GENETICS MUNKE, M., Cox, D. R., Jackson, I. J., HOGAN, B. L., FRANCKE, U. 1986; 42 (4): 236-240

    Abstract

    Two probes derived from a mouse recombinant lambda-clone (H24.1), that contains a sequence closely homologous to the Drosophila antennapedia homeo box, were mapped to mouse chromosome (MMU) 11 by filter hybridization of somatic cell hybrid DNA. This sequence is highly homologous to a human homeo box gene (HOX2) and appears to represent one of the two genes in the Hox-2 cluster previously assigned to MMU 11. To regionally map the Hox-2 cluster, we have carried out in situ hybridization of the two H24.1 probes and of an independently isolated Hox-2 probe. The autoradiographic silver grain distributions were similar in all three experiments with a peak over band 11D. This region contains the locus for the tail-short (Ts) mutation which causes skeletal abnormalities in heterozygotes and early embryonic death in homozygotes.

    View details for Web of Science ID A1986E458300011

    View details for PubMedID 2875852

  • STRUCTURAL FEATURES OF GROWTH-FACTOR RECEPTORS WITH ONCOGENIC POTENTIAL BIOCHEMICAL SOCIETY SYMPOSIA Chen, E., Coussens, L., Liao, Y. C., Smith, D., YANGFENG, T. L., McGrath, J., VANBEVEREN, C., Verma, I. M., Libermann, T. A., Schlessinger, J., FRANCKE, U., Levinson, A., Ullrich, A. 1986: 65-82

    View details for Web of Science ID A1986F939700006

    View details for PubMedID 3579970

  • CHROMOSOMAL MAPPING OF 4 DIFFERENT INTEGRATION SITES OF MOLONEY MURINE LEUKEMIA-VIRUS INCLUDING THE LOCUS FOR ALPHA-1(I) COLLAGEN IN MOUSE CYTOGENETICS AND CELL GENETICS MUNKE, M., Harbers, K., Jaenisch, R., FRANCKE, U. 1986; 43 (3-4): 140-149

    Abstract

    Infection of mouse embryos with Moloney murine leukemia virus (M-MuLV) has yielded several mouse substrains with stable germ line integration of retroviral DNA at distinct chromosomal loci (Mov loci; Jaenisch et al., 1981). There is evidence that flanking DNA sequences can have an effect on virus expression and, conversely, inserted viral DNA may affect the expression of adjacent host genes. As part of our studies on the interaction of inserted M-MuLV with the mouse genome, we have chromosomally mapped four different Mov loci by hybridizing single-copy mouse sequences, flanking the proviral DNA, to interspecies somatic cell hybrids. Furthermore, these sequences were assigned regionally by in situ hybridization to mouse metaphase chromosomes. In Mov-13 mice, M-MuLV had inserted into the alpha 1(I) collagen gene leading to early embryonic death in homozygotes. We have assigned this locus to the distal region of chromosome 11. Thus, the alpha 1(I) collagen gene is part of an evolutionarily conserved linkage group with the homologous genes on human chromosome 17. Three other proviral integration sites were mapped to chromosome 1, bands BC (Mov-7), chromosome 11, bands BC (Mov-9), and chromosome 3, bands FG (Mov-10). The Mov-10-specific probe detects an EcoRI-specific restriction fragment length polymorphism, which can make this probe a useful genetic marker.

    View details for Web of Science ID A1986G020900004

    View details for PubMedID 3467899

  • THE HUMAN C-REACTIVE PROTEIN GENE (CRP) AND SERUM AMYLOID-P COMPONENT GENE (APCS) ARE LOCATED ON THE PROXIMAL LONG ARM OF CHROMOSOME-1 IMMUNOGENETICS FLOYDSMITH, G., Whitehead, A. S., Colten, H. R., FRANCKE, U. 1986; 24 (3): 171-176

    Abstract

    The genes encoding two pentraxins, C-reactive protein (CRP) and serum amyloid P component (SAP), are located on the proximal long arm of human chromosome 1. Mapping of the CRP and SAP genes between the centromere and band q32 was achieved by Southern blot analysis of DNA from a panel of human X Chinese hamster somatic cell hybrids carrying defined fragments of human chromosome 1. Both genes were localized more precisely between bands q12 and q23 by in situ hybridization to human metaphase chromosomes.

    View details for Web of Science ID A1986E219700006

    View details for PubMedID 3759147

  • CHROMOSOMAL MAPPING OF GENES INVOLVED IN GROWTH-CONTROL COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY FRANCKE, U., YANGFENG, T. L., Brissenden, J. E., Ullrich, A. 1986; 51: 855-866

    View details for Web of Science ID A1986H050100017

    View details for PubMedID 3107886

  • REGIONAL ASSIGNMENTS OF 3 POLYMORPHIC DNA SEGMENTS ON HUMAN CHROMOSOME-15 GENETIC EPIDEMIOLOGY Brissenden, J. E., Page, D. C., DEMARTINVILLE, B., Trowsdale, J., Botstein, D., FRANCKE, U. 1986; 3 (4): 231-239

    Abstract

    Hybridization of probe pDP151 (locus D15S2) to genomic human DNAs digested with EcoRI revealed allelic restriction fragments 9 and 11 kilobase-pairs (kb) in length. Hybridization of pDP151 to EcoRI-digested DNAs from 21 Chinese hamster X human hybrid cell clones containing different subsets of human chromosomes demonstrated cosegregation of the 9 and 11 kb EcoRI fragments with human chromosome 15. D15S2 and two other polymorphic loci previously mapped to chromosome 15--D15S1 and D15S6--were localized to specific regions on human chromosome 15. Eight Chinese hamster X human somatic cell hybrid clones derived from a human donor heterozygous for a balanced translocation between chromosomes 15 and 22 [t(15;22)(q14;q13.3); Oliver et al, Cytogenet Cell Genet 22:503-510, 1978] were studied. After digestion of human and hybrid DNAs with HindIII and Southern blotting, pDP151 (D15S2) and pMS1-14 (D15S1) hybridized to fragments of 4 and 4.5 kb, respectively. Further, pMS1-14 (D15S1) and p9-1a (D15S6) hybridized to EcoRI fragments of 3.5 and 3.2 kb. All fragments cosegregated with the der(22) derivative chromosome containing region 15q14----15qter. In situ hybridization of these probes to normal human chromosomes mapped the corresponding loci with greater precision: D15S1 to 15q15----15q21, D15S2 to 15q15----15q22, and D15S6 to 15q22----15q24.

    View details for Web of Science ID A1986D806000003

    View details for PubMedID 3017806

  • HUMAN LUTEINIZING-HORMONE-RELEASING HORMONE GENE (LHRH) IS LOCATED ON SHORT ARM OF CHROMOSOME-8 (REGION 8P11.2-]P21) SOMATIC CELL AND MOLECULAR GENETICS YANGFENG, T. L., Seeburg, P. H., FRANCKE, U. 1986; 12 (1): 95-100

    Abstract

    Luteinizing hormone-releasing hormone (LHRH) is synthesized by hypothalamic neurons and affects the release of gonadotropic hormones from the anterior pituitary gland. A cDNA clone encoding the human LHRH precursor molecule was used to assign the LHRH gene to a human chromosome by in situ hybridization and Southern blot analysis. Metaphase spreads from two normal individuals were hybridized with 3H-labeled LHRH-specific sequence. Of 120 cells analyzed, 33 had silver grains over the p11.2----p21 bands of chromosome 8. No other chromosomal site was labeled above background, indicating the presence of a single site for LHRH sequences in the human genome. Independent confirmation for this location of the human LHRH gene on chromosome 8p was provided by analysis of DNA from human X Chinese hamster somatic cell hybrids. DNA samples were digested with EcoRI, blotted, and hybridized with the 32P-labeled human LHRH precursor cDNA probe. The single 11.5-kb human-specific band was detected only in hybrids containing human chromosome 8. Also, hybridization was observed in DNA from hybrids in which a portion of human chromosome 8 (region 8pter----8q21) had been spontaneously translocated onto a Chinese hamster chromosome.

    View details for Web of Science ID A1986A002700011

    View details for PubMedID 3511544

  • HUMAN GENES INVOLVED IN CHOLESTEROL-METABOLISM - CHROMOSOMAL MAPPING OF THE LOCI FOR THE LOW-DENSITY-LIPOPROTEIN RECEPTOR AND 3-HYDROXY-3-METHYLGLUTARYL-COENZYME-A REDUCTASE WITH CDNA PROBES PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Lindgren, V., LUSKEY, K. L., Russell, D. W., FRANCKE, U. 1985; 82 (24): 8567-8571

    Abstract

    Cellular cholesterol metabolism is regulated primarily through the coordinate expression of two proteins, the low density lipoprotein (LDL) receptor and 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase (EC 1.1.1.34). We have used cDNA probes for the human genes encoding these proteins to determine the precise chromosomal location of the two loci. By in situ hybridization we have regionally mapped the LDL receptor gene, LDLR, to the short arm of chromosome 19 in bands p13.1-p13.3. This result concurs with and extends a previous study in which LDLR was mapped to chromosome 19 by screening somatic cell hybrids with a species-specific monoclonal antibody. We have assigned the HMG-CoA reductase gene, HMGCR, to chromosome 5 by Southern blotting of DNA from a somatic cell hybrid panel and to bands 5q13.3-q14 by in situ hybridizations of the cDNA probe to human metaphase cells with normal and rearranged chromosomes.

    View details for Web of Science ID A1985AXL4200063

    View details for PubMedID 3866240

  • THE PRONATRIODILATIN GENE IS LOCATED ON THE DISTAL SHORT ARM OF HUMAN CHROMOSOME-1 AND ON MOUSE CHROMOSOME-4 AMERICAN JOURNAL OF HUMAN GENETICS YANGFENG, T. L., FLOYDSMITH, G., Nemer, M., Drouin, J., FRANCKE, U. 1985; 37 (6): 1117-1128

    Abstract

    Atrial natriuretic factors (ANF) are polypeptides having natriuretic, diuretic, and smooth muscle-relaxing activities that are synthesized from a single larger precursor: pronatriodilatin. Chromosomal assignment of the gene coding for human pronatriodilatin was accomplished by in situ hybridization of a [3H]-labeled pronatriodilatin probe to human chromosome preparations and by Southern blot analysis of somatic cell hybrid DNAs with normal and rearranged chromosomes 1. The human pronatriodilatin gene was mapped to the distal short arm of chromosome 1, in band 1p36. Southern blot analysis of mouse X Chinese hamster somatic cell hybrids was used to assign the mouse pronatriodilatin gene to chromosome 4. This assignment adds another locus to the conserved syntenic group of homologous genes located on the distal half of the short arm of human chromosome 1 and on mouse chromosome 4.

    View details for Web of Science ID A1985AXE3600008

    View details for PubMedID 2934979

  • TYROSINE KINASE RECEPTOR WITH EXTENSIVE HOMOLOGY TO EGF RECEPTOR SHARES CHROMOSOMAL LOCATION WITH NEU ONCOGENE SCIENCE Coussens, L., YANGFENG, T. L., Liao, Y. C., Chen, E., Gray, A., McGrath, J., Seeburg, P. H., Libermann, T. A., Schlessinger, J., FRANCKE, U., Levinson, A., Ullrich, A. 1985; 230 (4730): 1132-1139

    Abstract

    A novel potential cell surface receptor of the tyrosine kinase gene family has been identified and characterized by molecular cloning. Its primary sequence is very similar to that of the human epidermal growth factor receptor and the v-erbB oncogene product; the chromosomal location of the gene for this protein is coincident with the neu oncogene, which suggests that the two genes may be identical.

    View details for Web of Science ID A1985AUW2100010

    View details for PubMedID 2999974

  • HUMAN GENES FOR U2 SMALL NUCLEAR-RNA MAP TO A MAJOR ADENOVIRUS-12 MODIFICATION SITE ON CHROMOSOME-17 NATURE Lindgren, V., Ares, M., Weiner, A. M., FRANCKE, U. 1985; 314 (6006): 115-116

    Abstract

    U2 RNA is one of the abundant, highly conserved species of small nuclear RNA (snRNA) molecules implicated in RNA processing. As is typical of mammalian snRNAs, human U1 and U2 are each encoded by a multigene family. In the human genome, defective copies of the genes (pseudogenes) far outnumber the authentic genes. The majority or all of the 35 to 100 bona fide U1 genes have at least 20 kilobases (kb) of nearly perfect 5' and 3' flanking homology in common with each other; these U1 genes are clustered loosely in chromosome band 1p36 (refs 5, 7) with intergenic distances exceeding 44 kb. In contrast, the 10 to 20 U2 genes are clustered tightly in a virtually perfect tandem array which has a strict 6-kb repeating unit. We report here the assignment, by in situ hybridization, of the U2 gene cluster to chromosome 17, bands q21-q22. Surprisingly, this region is one of three major adenovirus 12 modification sites which undergo chromosome decondensation ('uncoiling') in permissive human cells infected by highly oncogenic strains of adenovirus. The two other major modification sites, 1p36 and 1q21, coincide with the locations of U1 genes and class I U1 pseudogenes, respectively. We suggest that snRNA genes are the major targets of viral chromosome modification.

    View details for Web of Science ID A1985ACV2800158

    View details for PubMedID 2579339

  • LOCALIZATION OF DNA-SEQUENCES IN REGION XP21 OF THE HUMAN X-CHROMOSOME - SEARCH FOR MOLECULAR MARKERS CLOSE TO THE DUCHENNE MUSCULAR-DYSTROPHY LOCUS AMERICAN JOURNAL OF HUMAN GENETICS DEMARTINVILLE, B., KUNKEL, L. M., Bruns, G., Morle, F., Koenig, M., Mandel, J. L., Horwich, A., LATT, S. A., Gusella, J. F., Housman, D., FRANCKE, U. 1985; 37 (2): 235-249

    Abstract

    Panels of somatic cell hybrid lines carrying various structural rearrangements of the human X chromosome short arm were analyzed with 21 X-chromosome-specific cloned DNA fragments. We mapped these molecular markers to five different regions of the short arm of the X chromosome. The results were confirmed by gene-dosage studies of human lymphoblasts with structurally abnormal X chromosomes. The ornithine transcarbamylase gene and four anonymous DNA sequences map within band Xp21, flanking the presumed locus for Duchenne muscular dystrophy.

    View details for Web of Science ID A1985AGY5900001

    View details for PubMedID 2984924

  • MINUTE CHROMOSOMES REPLACING THE Y-CHROMOSOME CARRY Y-SPECIFIC SEQUENCES BY RESTRICTION FRAGMENT ANALYSIS AND INSITU HYBRIDIZATION AMERICAN JOURNAL OF MEDICAL GENETICS MUNKE, M., DEMARTINVILLE, B., Lieber, E., FRANCKE, U. 1985; 22 (2): 361-374

    Abstract

    Two unrelated males, a 43-year-old man with azoospermia and a 4-year-old boy with stature at the 10th centile, had similar karyotypes: 46,X,min. The minutes, present in all cells analyzed, stained weakly with G-, C-, and Q-banding methods. To elucidate their origin we used molecular techniques: In HaeIII digests of total genomic DNA from both individuals, no Y-specific reiterated sequences were detected. However, restriction fragment analysis with probe pDP31 demonstrated that the patients' DNA contained the Y-specific fragment. In situ hybridization with the same probe showed that these sequences were present on the minute chromosomes and have not been translocated elsewhere.

    View details for Web of Science ID A1985AQL3500020

    View details for PubMedID 2996350

  • MAPPING OF TRANSFORMING GROWTH-FACTOR ALPHA-GENE ON HUMAN CHROMOSOME-2 CLOSE TO THE BREAKPOINT OF THE BURKITTS-LYMPHOMA T(2,8) VARIANT TRANSLOCATION CANCER RESEARCH Brissenden, J. E., DERYNCK, R., FRANCKE, U. 1985; 45 (11): 5593-5597

    Abstract

    Transforming growth factors (TGFs) are defined as biologically active polypeptides which reversibly confer the transformed phenotype onto untransformed cultured cells. TGF-alpha shows sequence homology with epidermal growth factor and competes with epidermal growth factor for binding to the epidermal growth factor receptor, stimulating the phosphorylation of the receptor. TGF-alpha is secreted by many transformed cells and may be involved in embryonic development. A cloned human TGF-alpha gene was used to map the locus for the TGF-alpha precursor to the short arm of human chromosome 2, region 2p11----2p13, by Southern blotting techniques with DNA prepared from rodent X human somatic cell hybrids. These hybrids contained different subsets of human chromosomes and included a set with a translocation between human chromosomes 1 and 2 [t(1;2) (q32;q13)]. In situ hybridization of the TGF-alpha probe to normal human metaphase spreads confirmed these data and localized TGF-alpha more precisely to bands 2p11----2p13. Breakpoints in the variant Burkitt lymphoma translocation t(2;8) occur within these bands. Such a t(2;8) translocation could place TGF-alpha next to c-myc in band 8q24. The possibility is raised that TGF-alpha might contribute to tumor progression in these cases of Burkitt's lymphoma.

    View details for Web of Science ID A1985ATM3000003

    View details for PubMedID 3863708

  • ASSIGNMENT OF THE HUMAN-GENE FOR THE GLUCOCORTICOID RECEPTOR TO CHROMOSOME-5 PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Gehring, U., SEGNITZ, B., FOELLMER, B., FRANCKE, U. 1985; 82 (11): 3751-3755

    Abstract

    Human lymphoblastic leukemia cells of line CEM-C7 are glucocroticoid-sensitive and contain glucocorticoid receptors of wild-type characteristics. EL4 mouse lymphoma cells are resistant to lysis by glucocorticoids due to mutant receptors that exhibit abnormal DNA binding. Hybrids between the two cell lines were prepared and analyzed with respect to glucocorticoid responsiveness and to receptor types by DNA-cellulose chromatrography. Sensitive hybrid cell clones contained the CEM-C7-specific receptor in addition to the EL4 type of receptor. Several sensitive hybrid cell clones were used for selection of resistant segregants by growth in the presence of high concentrations of glucocorticoid. These segregants had lost the wild-type CEM-C7 receptor, while the EL4-specific receptor was retained. To identify the human chromosome that was lost concordantly with the CEM-C7 receptor the chromosomes of hybrid cells were studied by alkaline Giemsa (G-11) staining and trypsin/Giemsa banding. All hybrids contained human chromosomes in addition to one to two sets of EL4 chromosomes. Human chromosome 5 was present in all hybrid cell clones that expressed the CEM-C7 receptor and it was absent from those that did not. This absolute correlation was not observed for any other human chromosome. We conclude that the human gene for the glucocorticoid receptor is located on chromosome 5.

    View details for Web of Science ID A1985AKB7900048

    View details for PubMedID 3858847

  • HUMAN-U-1 SMALL NUCLEAR-RNA PSEUDOGENES DO NOT MAP TO THE SITE OF THE U-1 GENES IN 1P36 BUT ARE CLUSTERED IN 1Q12-Q22 MOLECULAR AND CELLULAR BIOLOGY Lindgren, V., BERNSTEIN, L. B., Weiner, A. M., FRANCKE, U. 1985; 5 (9): 2172-2180

    Abstract

    Human U1 small nuclear RNA is encoded by approximately 30 gene copies. All of the U1 genes share several kilobases of essentially perfect flanking homology both upstream and downstream from the U1 coding region, but remarkably, for many U1 genes excellent flanking homology extends at least 24 kilobases upstream and 20 kilobases downstream. Class I U1 RNA pseudogenes are abundant in the human genome. These pseudogenes contain a complete but imperfect U1 coding region and possess extensive flanking homology to the true U1 genes. We mapped four class I pseudogenes by in situ hybridization to the long arm of chromosome 1, bands q12-q22, a region distinct from the site on the distal short arm of chromosome 1 to which the U1 genes have been previously mapped (Lund et al., Mol. Cell. Biol. 3:2211-2220, 1983; Naylor et al., Somat. Cell Mol. Genet. 10:307-313, 1984). We confirmed our in situ hybridization results by genomic blotting experiments with somatic cell hybrid lines with translocation products of human chromosome 1. These experiments provide further evidence that class I U1 pseudogenes and the true U1 genes are not interspersed. The results, along with those published elsewhere (Bernstein et al., Mol. Cell. Biol. 5:2159-2171, 1985), suggest that gene amplification may be responsible for the sequence homogeneity of the human U1 gene family.

    View details for Web of Science ID A1985AQH5200002

    View details for PubMedID 3837186

  • HIGH-RESOLUTION CYTOGENETIC EVALUATION OF COUPLES WITH RECURRING FETAL WASTAGE HUMAN GENETICS YANGFENG, T. L., Finley, S. C., Finley, W. H., FRANCKE, U. 1985; 69 (3): 246-249

    Abstract

    Both high resolution and routine chromosome analyses were used to study couples with a history of two or more spontaneous abortions in early pregnancy. In the 20 couples studied, two of the women were found to have an inversion. One paracentric inversion, (13)(q13.1q22.3), was apparent on routine analysis. The small pericentric inversion (11)(p11.12q12.3), was only detected by high resolution techniques. Given the low yield and increased cost and effort involved, we do not believe that high resolution studies are justified for screening couples with repeated abortions.

    View details for Web of Science ID A1985AFZ0300012

    View details for PubMedID 3980017

  • THE GENE ENCODING THE IA-ASSOCIATED INVARIANT CHAIN IS LOCATED ON CHROMOSOME-18 IN THE MOUSE IMMUNOGENETICS Yamamoto, K., FLOYDSMITH, G., FRANCKE, U., Koch, N., Lauer, W., Dobberstein, B., SCHAFER, R., Hammerling, G. J. 1985; 21 (1): 83-90

    Abstract

    The chromosomal assignment of the gene encoding the invariant (Ii) chain associated with the mouse immune response antigens (Ia) was determined by Southern blot analysis of DNA from a panel of mouse X Chinese hamster somatic cell hybrids cleaved with Hind III or Eco RI. Using a mouse li cDNA as a hybridization probe, we localized the gene coding for the invariant chain to mouse chromosome 18.

    View details for Web of Science ID A1985AAS7400009

    View details for PubMedID 2981770

  • DIRECT ASSIGNMENT OF OROSOMUCOID TO HUMAN CHROMOSOME-9 AND ALPHA-2HS-GLYCOPROTEIN TO CHROMOSOME-3 USING HUMAN-FETAL LIVER-X RAT HEPATOMA HYBRIDS HUMAN GENETICS Cox, D. W., FRANCKE, U. 1985; 70 (2): 109-115

    Abstract

    The production of plasma proteins has been monitored in somatic cell hybrids between a rat hepatoma cell line (7777) and human fetal liver cells. Production of 14 plasma proteins was assayed in concentrated serum-free culture supernatants by electroimmunoassay. Alpha 2HS-glycoprotein (AHSG) was produced by 10 of 19 hybrids; concordancy for presence or absence of protein production was 100% for human chromosome 3. Orosomucoid (ORM) was produced in 8 of 19 hybrids, with a concordancy for presence or absence of protein of 94.7% with human chromosome 9. The chromosome location for genes for these two proteins, previously assigned by linkage studies, is confirmed by direct assignment. These studies have also suggested possible chromosomal assignments for loci for alpha 1-antichymotrypsin and C1 esterase inhibitor. Other genes for proteins which could not be assigned to specific chromosomes using these hybrids were: complement C3, ceruloplasmin, hemopexin, inter-alpha-trypsin inhibitor, prealbumin, retinol-binding protein, transferrin and apolipoproteins CII, B, and sinking-pre-beta [Lp(a)].

    View details for Web of Science ID A1985AKU8900002

    View details for PubMedID 3859464

  • ASSIGNMENT OF THE STRUCTURAL GENE FOR SUBUNIT-M1 OF HUMAN RIBONUCLEOTIDE REDUCTASE TO THE SHORT ARM OF CHROMOSOME-11 EXPERIMENTAL CELL RESEARCH Engstrom, Y., FRANCKE, U. 1985; 158 (2): 477-483

    Abstract

    By using a species-specific monoclonal antibody that recognizes subunit M1 of ribonucleotide reductase from human but not hamster origin, we have been able to assign the structural gene for the human protein M1 to the short arm of chromosome 11. Protein extracts from a panel of human-Chinese hamster somatic cell hybrids were subjected to electrophoresis in sodium dodecyl sulfate (SDS) denaturating polyacrylamide gels, and then transferred and coupled covalently to diazobenzyloxymethyl paper. These were screened for human protein M1 by incubation with the mouse monoclonal anti-M1 antibody AD 203, followed by rabbit anti-mouse IgG, 125I-labelled Staphylococcus protein A and finally autoradiography. In all tested hybrids the detection of human protein M1 was correlated with the presence of chromosome 11, specifically with the short arm of this chromosome. This region also contains the human genes for insulin, insulin-like growth factor II, and the c-Harvey-ras 1 oncogene.

    View details for Web of Science ID A1985ALZ4100017

    View details for PubMedID 3891388

  • GENE FOR HUMAN INSULIN-RECEPTOR - LOCALIZATION TO SITE ON CHROMOSOME-19 INVOLVED IN PRE-B-CELL LEUKEMIA SCIENCE YANGFENG, T. L., FRANCKE, U., Ullrich, A. 1985; 228 (4700): 728-731

    Abstract

    Consistent chromosomal translocations in neoplastic cells may alter the expression of proto-oncogenes that are located near the breakpoints. The complementary DNA sequence of the human insulin receptor is similar to those of the EGF receptor (erbB oncogene) and products of the src family of oncogenes. With in situ hybridization and Southern blot analysis of somatic cell hybrid DNA, the human insulin receptor gene was mapped to the distal short arm of chromosome 19 (bands p13.2----p13.3), a site involved in a nonrandom translocation in pre-B-cell acute leukemia.

    View details for Web of Science ID A1985AGH4200033

    View details for PubMedID 3873110

  • THE NEU GENE - AN ERBB-HOMOLOGOUS GENE DISTINCT FROM AND UNLINKED TO THE GENE ENCODING THE EGF RECEPTOR SCIENCE SCHECHTER, A. L., Hung, M. C., Vaidyanathan, L., Weinberg, R. A., YANGFENG, T. L., FRANCKE, U., Ullrich, A., Coussens, L. 1985; 229 (4717): 976-978

    Abstract

    The neu oncogene, identified in ethylnitrosourea-induced rat neuroglioblastomas, had strong homology with the erbB gene that encodes the epidermal growth factor receptor. This homology was limited to the region of erbB encoding the tyrosine kinase domain. It was concluded that the neu gene is a distinct novel gene, as it is not coamplified with sequences encoding the EGF receptor in the genome of the A431 tumor line and it maps to human chromosome 17.

    View details for Web of Science ID A1985APR0600030

    View details for PubMedID 2992090

  • MINOR XP21 CHROMOSOME DELETION IN A MALE ASSOCIATED WITH EXPRESSION OF DUCHENNE MUSCULAR-DYSTROPHY, CHRONIC GRANULOMATOUS-DISEASE, RETINITIS PIGMENTOSA, AND MCLEOD SYNDROME AMERICAN JOURNAL OF HUMAN GENETICS FRANCKE, U., Ochs, H. D., DEMARTINVILLE, B., Giacalone, J., Lindgren, V., Disteche, C., Pagon, R. A., Hofker, M. H., VANOMMEN, G. J., Pearson, P. L., WEDGWOOD, R. J. 1985; 37 (2): 250-267

    Abstract

    We are reporting a male patient who suffered from chronic granulomatous disease associated with cytochrome b-245 deficiency and McLeod red cell phenotype, Duchenne muscular dystrophy, and retinitis pigmentosa. On cytogenetic analysis, he seemed to have a very subtle interstitial deletion of part of band Xp21. Since it was impossible to know whether this material was truly deleted or inserted elsewhere in the genome, somatic cell and molecular studies were carried out. In somatic cell hybrids, the deleted X chromosome was isolated on a Chinese hamster background. Southern blot analysis with 20 single-copy probes, that had been mapped to the X short arm, led to the discovery of one (probe 754) that is missing from this patient's X chromosome and also from his total DNA. This proves that he, indeed, has a deletion rather than a balanced insertion. The results provide cytological mapping information for the X-linked phenotypes present in this patient. Furthermore, probe 754 recognizes a restriction fragment length polymorphism of high frequency that makes it the most powerful probe currently available for linkage studies with X-linked muscular dystrophy.

    View details for Web of Science ID A1985AGY5900002

    View details for PubMedID 4039107

  • AMPLIFIED DNA IN Y1 MOUSE ADRENAL-TUMOR CELLS - ISOLATION OF CDNAS COMPLEMENTARY TO AN AMPLIFIED C-KI-RAS GENE AND LOCALIZATION OF HOMOLOGOUS SEQUENCES TO MOUSE CHROMOSOME-6 NUCLEIC ACIDS RESEARCH George, D. L., Scott, A. F., DEMARTINVILLE, B., FRANCKE, U. 1984; 12 (6): 2731-2743

    Abstract

    We have isolated cDNA clones complementary to a c-Ki-ras cellular oncogene that is amplified in Y1 mouse adrenal tumor cells, with the amplified sequences located on double-minute chromatin bodies (DMs) and homogeneously staining chromosomal regions (HSRs). Characterization of the cDNAs included the isolation of corresponding genomic clones, Northern blot analysis of RNA, and DNA sequence analysis. Our studies demonstrate that the c-Ki-ras gene amplified in the Y1 cells is homologous to the human c-Ki-ras2 gene. We have also obtained evidence that, in addition to c-Ki-ras, at least one other transcription unit has been amplified in the mouse adrenal tumor cells. Moreover, by Southern blot analysis of Chinese hamster-mouse somatic cell hybrids, we have determined that the amplified DNA sequences associated with DMs and HSRs, including the c-Ki-ras gene, are present in normal mouse cells on chromosome 6.

    View details for Web of Science ID A1984SK10500011

    View details for PubMedID 6546797

  • ASSIGNMENT OF THE STRUCTURAL GENE ENCODING HUMAN ASPARTYLGLUCOSAMINIDASE TO THE LONG ARM OF CHROMOSOME-4 (4Q21-]4QTER) AMERICAN JOURNAL OF HUMAN GENETICS AULA, P., Astrin, K. H., FRANCKE, U., Desnick, R. J. 1984; 36 (6): 1215-1224

    Abstract

    The structural gene for the human lysosomal enzyme aspartylglucosaminidase (AGA) has been assigned to chromosome 4 using somatic cell hybridization techniques. The human monomeric enzyme was detected in Chinese hamster-human cell hybrids by a thermal denaturation assay that selectively inactivated the Chinese hamster isozyme, while the thermostable human enzyme retained activity. Twenty informative hybrid clones, derived from seven independent fusions, were analyzed for the presence of human AGA activity and their human chromosomal constitutions. Without exception, the presence of human AGA in these hybrids was correlated with the presence of human chromosome 4. All other human chromosomes were excluded by discordant segregation of the human enzyme and other chromosomes. Two hybrid clones, with interspecific Chinese hamster-human chromosome translocations involving the long arm of human chromosome 4, permitted the assignment of human AGA to the region 4q21----4qter.

    View details for Web of Science ID A1984TZ59000006

    View details for PubMedID 6517050

  • RANDOM-X INACTIVATION RESULTING IN MOSAIC NULLISOMY OF REGION XP21.1-]P21.3 ASSOCIATED WITH HETEROZYGOSITY FOR ORNITHINE TRANSCARBAMYLASE DEFICIENCY AND FOR CHRONIC GRANULOMATOUS-DISEASE CYTOGENETICS AND CELL GENETICS FRANCKE, U. 1984; 38 (4): 298-307

    Abstract

    A young woman with normal gonadal development and mild mental retardation was found to have a small de novo interstitial deletion of most of band Xp21, karyotype designation 46,X,del(X) (pter----p21.3:: p21.1----qter). Replication studies on lymphocytes and skin fibroblasts revealed that in 45% of cells the normal X was late replicating. Somatic cell hybrids between her fibroblasts and HPRT-deficient Chinese hamster cells were obtained and selected for and against retention of the active human X chromosome. In several independent hybrids the deleted X was retained in the active state. Partial ornithine transcarbamylase (ornithine carbamoyltransferase EC 2.1.3.3) (OTC) deficiency was documented by elevated urinary orotic acid excretion and increased serum glutamine after a protein load. This confirms the mapping of the structural gene for OTC to this deletion. Testing of neutrophil function revealed heterozygosity for chronic granulomatous disease (CGD) suggesting that a gene for CGD maps within the deletion. Thus, X inactivation mosaicism is also present in hepatocytes and neutrophilic granulocytes. Random X inactivation in a female with an Xp deletion has not been previously reported. The cells from this patient and the somatic cell hybrids containing her deleted X chromosome in the absence of the normal X provide material for the precise mapping of X linked genes and DNA sequences on the short arm of the human X chromosome.

    View details for Web of Science ID A1984TW47100009

    View details for PubMedID 6510024

  • DNA ANALYSIS OF 1ST-TRIMESTER CHORIONIC VILLOUS BIOPSIES - TEST FOR MATERNAL CONTAMINATION AMERICAN JOURNAL OF HUMAN GENETICS DEMARTINVILLE, B., BLAKEMORE, K. J., Mahoney, M. J., FRANCKE, U. 1984; 36 (6): 1357-1368

    Abstract

    We investigated the reliability of chorionic villous biopsy as a method to obtain tissues reflecting the genetic constitution of the embryo. In 12 pregnancies before elective termination, we searched for detectable maternal tissue after careful dissection of villi from small 2-5-mg specimens that yielded 7 micrograms of DNA per mg tissue. In Southern blotting experiments (1-2 micrograms DNA per lane), restriction fragment length polymorphisms (RFLPs) at an autosomal (D14S1) and a sex chromosomal (DXYS1) locus allowed recognition of maternally and embryonically derived alleles. Pure villi were obtained in six of the seven informative cases. One biopsy was not dissected satisfactorily; a mixture of embryonic and maternal DNA was found. Nonvillous tissues were mostly maternally derived in eight informative cases. Sex determination by molecular analysis (alleles at the DXYS1 locus) agreed with the karyotypes of uncultured or cultured villi. In one continuing pregnancy, distinct RFLPs indicated maternal inheritance of the alpha-thalassemia 1 trait in a female embryo without detectable maternal contamination. Reliable prenatal diagnosis depends on the specimen's purity. Maternal contamination can be evaluated by DNA analyses.

    View details for Web of Science ID A1984TZ59000017

    View details for PubMedID 6517057

  • HUMAN ORNITHINE TRANSCARBAMYLASE LOCUS MAPPED TO BAND XP21.1 NEAR THE DUCHENNE MUSCULAR-DYSTROPHY LOCUS SCIENCE Lindgren, V., DEMARTINVILLE, B., Horwich, A. L., Rosenberg, L. E., FRANCKE, U. 1984; 226 (4675): 698-700

    Abstract

    The gene for the mitochondrial enzyme ornithine transcarbamylase was mapped to the short arm of the X chromosome by in situ hybridization experiments, with DNA complementary to the human ornithine transcarbamylase gene used as a probe. A series of cell lines with X chromosome abnormalities was used to localize the gene to band Xp21.1. Because the gene maps near the Duchenne muscular dystrophy locus, the ornithine transcarbamylase probe may be useful in carrier detection and prenatal diagnosis of Duchenne muscular dystrophy as well as of ornithine transcarbamylase deficiency.

    View details for Web of Science ID A1984TQ42500035

    View details for PubMedID 6494904

  • ASSIGNMENT OF THE GENE FOR CYSTATHIONINE BETA-SYNTHASE TO HUMAN CHROMOSOME-21 IN SOMATIC-CELL HYBRIDS HUMAN GENETICS Skovby, F., KRASSIKOFF, N., FRANCKE, U. 1984; 65 (3): 291-294

    Abstract

    Among several established mouse, rat, and Chinese hamster cell lines that were screened for cystathionine beta-synthase (CBS) activity, mouse 3T3 and Chinese hamster Don fibroblasts were found to contain no detectable activity. Somatic cell hybrids between human fibroblasts KG-7 with normal CBS activity and Don/a23TK- cells (series XXI) were examined for CBS activity and for human chromosome content. Only chromosome 21 cosegregated with CBS activity. Because the activities measured could represent either Chinese hamster or human gene products, we have prepared a new series of hybrids between Don/a23TK- cells and mutant human fibroblasts from a patient with homocystinuria due to deficiency of functional CBS mRNA. None of these (series XXV) hybrids contained detectable CBS activity, although collectively all human chromosomes were represented. Our results suggest that the human gene for CBS, called CBS, and thus for the most common form of homocystinuria, is located on chromosome 21.

    View details for Web of Science ID A1984SD45400014

    View details for PubMedID 6583157

  • ASSIGNMENT OF THE HUMAN-GENE FOR THE LOW-DENSITY LIPOPROTEIN RECEPTOR TO CHROMOSOME-19 - SYNTENY OF A RECEPTOR, A LIGAND, AND A GENETIC-DISEASE PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA-BIOLOGICAL SCIENCES FRANCKE, U., BROWN, M. S., Goldstein, J. L. 1984; 81 (9): 2826-2830

    Abstract

    The availability of a species-specific monoclonal antibody that recognizes the low density lipoprotein (LDL) receptor of human but not hamster origin permitted assignment of the structural gene for the human receptor to chromosome 19. The antibody was used to detect the human LDL receptor in a series of hamster-human somatic cell hybrids by two assays: (i) a structural assay that measured cellular incorporation of [35S]methionine into immunoprecipitable receptor and (ii) a functional assay that measured the rate of receptor-dependent uptake and degradation of the 125I-labeled anti-receptor monoclonal antibody. Both assays showed that the human LDL receptor was expressed in 15 out of 20 hybrid cell lines. Expression of the human LDL receptor was 100% concordant with the presence of human chromosome 19; all other human chromosomes showed at least 25% discordance. As expected, the gene for the LDL receptor (LDLR) is located on the same chromosome as the gene for the disease familial hypercholesterolemia, which has been previously mapped to chromosome 19 by pedigree studies and is caused by allelic mutations at the LDL receptor locus. The gene for apolipoprotein E, a ligand for the LDL receptor, is also known to be located on chromosome 19, raising the possibility of an evolutionary link between a protein ligand and its receptor.

    View details for Web of Science ID A1984SR73200047

    View details for PubMedID 6326146

  • HUMAN CHROMOSOMAL MAPPING OF GENES FOR INSULIN-LIKE GROWTH FACTOR-I AND FACTOR-II AND EPIDERMAL GROWTH-FACTOR NATURE Brissenden, J. E., Ullrich, A., FRANCKE, U. 1984; 310 (5980): 781-784

    Abstract

    Many of the actions previously attributed to pituitary-derived growth hormone are mediated by polypeptide growth factors. These include the insulin-like growth factors I and II (IGF-I and IGF-II), which are members of the insulin family of proteins. We report here the chromosomal mapping of the human genes for IGF-I and IGF-II. IGF-II maps to the short arm of chromosome 11, which also contains the gene for insulin and the proto-oncogene c-Ha-ras1 (ref. 9). IGF-I maps to chromosome 12, which is evolutionarily related to chromosome 11 and carries the gene for the proto-oncogene c-Ki-ras2 (refs 10,44). We have also localized the human gene for an unrelated polypeptide hormone, epidermal growth factor, to chromosome 4q, in the same region as another specialized growth factor, T-cell growth factor. We speculate that these map assignments reflect the existence of gene families involved in growth control.

    View details for Web of Science ID A1984TG79200046

    View details for PubMedID 6382023

  • COMPARATIVE-ANALYSIS OF MOUSE HUMAN HYBRIDS WITH REARRANGED CHROMOSOMES-1 BY INSITU HYBRIDIZATION AND SOUTHERN BLOTTING - HIGH-RESOLUTION MAPPING OF NRAS, NGFB, AND AMY ON HUMAN CHROMOSOME-1 SOMATIC CELL AND MOLECULAR GENETICS MUNKE, M., Lindgren, V., DEMARTINVILLE, B., FRANCKE, U. 1984; 10 (6): 589-599

    Abstract

    The human protooncogene NRAS and the genes for the beta-subunit of nerve growth factor (NGFB) and for amylase (AMY) have previously been assigned to the proximal short arm of chromosome 1, but their precise positions have not been unequivocally established. By in situ hybridization of DNA probes for the three genes, we have ascertained the location of complementary sequences in mouse-human somatic cell hybrids that contained translocations of chromosome 1. The results agreed with the presence or absence of the human sequences as determined by Southern blotting of hybrid cell DNA. The in situ data confirmed that the genes were present on the cytologically recognized rearranged chromosome. Compared to the autoradiographic silver grain distribution on normal human chromosome 1, our in situ results obtained with the translocation chromosomes allowed much greater precision of mapping. Both NRAS and NGFB map to band 1p22, and AMY was confirmed in band 1p21.

    View details for Web of Science ID A1984TW24800005

    View details for PubMedID 6209808

  • Chromosomal assignments of three random RFLP loci defined by base-pair changes in MspI sites. Molecular biology & medicine de Martinville, B., Schäfer, M., White, R., FRANCKE, U. 1983; 1 (4): 415-424

    Abstract

    Three cloned single copy sequences isolated from a random partial plasmid library of human EcoRI-HindIII restriction fragments detect MspI site polymorphisms in human DNA. Since at least two of them have allele frequencies that make them suitable for family linkage studies, we have determined their chromosomal localizations. DNA samples extracted from a panel of 31 Chinese hamster x human somatic cell hybrids, that were derived from six different human donors, were analyzed for the presence of human EcoRI or HindIII fragments hybridizing with the three probes. The results were correlated with the human chromosome contents of the hybrids. Concordancy was observed for clone pMS 1-14 (preliminary locus designation DOSLC3) and human chromosome 15, for clone pMS 1-27 (DOSLC2) and chromosome 20, and for clone pMS 1-37 (DOSLC1) and chromosome 3. Two independent hybrids had retained only the long arm of chromosome 3 and did not hybridize with this probe. Therefore, we assign this locus (now termed D3S3) to the short arm of chromosome 3.

    View details for PubMedID 6209528

  • Brief clinical report: aqueductal stenosis leading to hydrocephalus--an unusual manifestation of neurofibromatosis. American journal of medical genetics Horwich, A., Riccardi, V. M., FRANCKE, U. 1983; 14 (3): 577-581

    Abstract

    Two brothers and an unrelated male with neurofibromatosis (NF) developed hydrocephalus in childhood. Aqueductal stenosis (AS) was demonstrated by ventriculography and required shunt operation. Genetic counselors had to decide whether to invoke an X-linked gene for AS or whether AS could be caused by the NF mutation. Reports of 13 patients of both sexes with AS and NF suggest that AS with resulting hydrocephalus may be a rare manifestation of the NF gene. AS should be looked for in young NF patients with signs of hydrocephalus or with neurologic abnormalities.

    View details for PubMedID 6407319

  • THE N-RAS ONCOGENE ASSIGNED TO THE SHORT ARM OF HUMAN CHROMOSOME-1 NUCLEIC ACIDS RESEARCH DEMARTINVILLE, B., Cunningham, J. M., Murray, M. J., FRANCKE, U. 1983; 11 (15): 5267-5275

    Abstract

    The human N-ras oncogene, isolated from the HL-60 promyelocytic leukemia cell line, is distantly related to viral oncogenes of Kirsten and Harvey sarcoma viruses. We have determined its chromosomal location by Southern blot analysis of DNAs from 37 human x rodent hybrid cell lines derived from 8 different human donors, some of whom carried balanced rearrangements of chromosome 1. The results indicate that the N-ras oncogene (RASN) is localized on the proximal part of the short arm of human chromosome 1, in region p3200 leads to cen.

    View details for Web of Science ID A1983RC32300022

    View details for PubMedID 6576326

  • THE C-HA-RAS1, INSULIN AND BETA-GLOBIN LOCI MAP OUTSIDE THE DELETION ASSOCIATED WITH ANIRIDIA-WILMS TUMOR NATURE DEMARTINVILLE, B., FRANCKE, U. 1983; 305 (5935): 641-643

    Abstract

    The localization of protooncogenes on human chromosomes may coincide with chromosome breakpoints of consistent translocations in leukaemias or lymphomas, suggesting a direct involvement of oncogenes in carcinogenesis. For example, in Burkitt's lymphoma consistent translocations may be associated with rearrangements of c-myc. Our assignment of the c-Harvey-ras1 oncogene to chromosome 11, precisely to region 11p11 leads to p15 (ref. 5; not 11p13 as stated in ref. 6), has raised the possibility that this oncogene might have a role in the predisposition to nephroblastoma (Wilms' tumour, WT) seen in the aniridia-WT association (AWTA) that is frequently caused by an interstitial deletion of band 11p (ref. 8). We have now studied the organization and copy number of sequences at three loci mapped to 11p: c-Ha-ras1, insulin and gamma-globin in cells from four individuals with structural rearrangements of the short arm of chromosome 11. Our results reported here rule out a close physical linkage between c-Ha-ras1 and the genes responsible for AWTA, and suggest a more distal localization of the beta-globin cluster than currently assumed.

    View details for Web of Science ID A1983RL31400058

    View details for PubMedID 6312329

  • REGIONAL ASSIGNMENT OF THE HUMAN-GENE FOR PLATELET-TYPE PHOSPHOFRUCTOKINASE (PFKP) TO CHROMOSOME-10P - NOVEL USE OF POLYSPECIFIC RODENT ANTISERA TO LOCALIZE HUMAN ENZYME GENES HUMAN GENETICS Vora, S., Miranda, A. F., Hernandez, E., FRANCKE, U. 1983; 63 (4): 374-379

    Abstract

    Human phosphofructokinase (PFK; EC 2.7.1.11) is under the control of three structural loci which encode muscle-type (M), liver-type (L), and platelet or fibroblast-type (P) subunits; human diploid fibroblasts and leukocytes express all three loci. In order to assign the human PFKP locus to a specific human chromosome, in this study, we have examined ten human X rodent somatic cell hybrids for the expression of human P subunits using a mouse anti-human P subunit-specific antiserum in an active-enzyme-immunoprecipitation technique. In nine of ten hybrids studied, the expression of the PFKP locus segregated concordantly with chromosome 10 and none other, indicating that PFKP is located on chromosome 10; the discordancy rates for all the other chromosomes were 0.2 or greater. In the one discordant hybrid, only the long arm of chromosome 10 was retained and PFKP was not expressed. Human fibroblasts from a patient with duplication of the short arm of chromosome 10 consistently exhibited PFK activity values 180% of normal. These data indicate that human PFKP is located on the short arm of chromosome 10, and that a gene dosage effect is demonstrable in fibroblasts with a duplication of 10p. The use of rodent antihuman antibody combined with immunoprecipitation aided by staphylococci-bearing protein A may find general application in mapping human enzyme genes, when human and rodent gene-products are not distinguishable by other means.

    View details for Web of Science ID A1983QY02700016

    View details for PubMedID 6222962

  • CHROMOSOME MAPPING OF HUMAN CELL-SURFACE MOLECULES - MONOCLONAL ANTI-HUMAN LYMPHOCYTE ANTIBODY-4F2, ANTIBODY-A3D8, AND ANTIBODY-A1G3 DEFINE ANTIGENS CONTROLLED BY DIFFERENT REGIONS OF CHROMOSOME-11 SOMATIC CELL GENETICS FRANCKE, U., FOELLMER, B. E., HAYNES, B. F. 1983; 9 (3): 333-344

    Abstract

    Monoclonal antibodies 4F2, A3D8, and A1G3, directed against cell surface antigens present on subsets of human cells, were used to identify the human chromosome regions that code for the antigenic determinants. Human fibroblasts expressed all three antigens, and no cross-reactivity with Chinese hamster or mouse cells was found. Fourteen rodent X human somatic cell hybrids, derived from six different human donors and from two different Chinese hamster and one mouse cell line, were studied simultaneously for human chromosome content and for antibody binding as detected by indirect immunofluorescence. Concordancy with binding of all three antibodies was observed only for human chromosome 11. All other chromosomes were excluded by three or more discordant hybrid clones. Data from six hybrids containing three different regions of chromosome 11 indicate that it is the long arm of chromosome 11 which is both necessary and sufficient for expression of the human antigen defined by 4F2 while the antigen(s) defined by A3D8 and A1G3 map to short arm.

    View details for Web of Science ID A1983QQ50600003

    View details for PubMedID 6190235

  • ONCOGENE FROM HUMAN EJ BLADDER-CARCINOMA IS LOCATED ON THE SHORT ARM OF CHROMOSOME-11 SCIENCE DEMARTINVILLE, B., Giacalone, J., Shih, C., Weinberg, R. A., FRANCKE, U. 1983; 219 (4584): 498-501

    Abstract

    The human cellular homolog of the transforming DNA sequence isolated from the bladder carcinoma cell line EJ was localized on the short arm of human chromosome 11 by Southern blot analysis of human-rodent hybrid cell DNA. This locus contains human sequences homologous to the Harvey murine sarcoma virus v-Ha-ras oncogene.

    View details for Web of Science ID A1983PZ52300021

    View details for PubMedID 6297001

  • THE HUMAN-GENE FOR THE BETA-SUBUNIT OF NERVE GROWTH-FACTOR IS LOCATED ON THE PROXIMAL SHORT ARM OF CHROMOSOME-1 SCIENCE FRANCKE, U., DEMARTINVILLE, B., Coussens, L., Ullrich, A. 1983; 222 (4629): 1248-1251

    Abstract

    Fragments of the recently cloned human gene for the beta subunit of nerve growth factor (beta-NGF) were used as hybridization probes in analyzing two sets of rodent-human somatic cell hybrids for the presence of human beta-NGF sequences. Results from the first set of hybrids assigned the human beta-NGF gene to chromosome 1 and ruled out the presence of sequences of comparable homology on any other chromosome. With the second set of hybrids, which contained seven different, but overlapping, regions of chromosome 1, the NGF locus was mapped to band 1p22.

    View details for Web of Science ID A1983RT72700033

    View details for PubMedID 6648531

  • FUSION OF DNA REGION TO MURINE IMMUNOGLOBULIN HEAVY-CHAIN LOCUS CORRESPONDS TO PLASMACYTOMA-ASSOCIATED CHROMOSOME-TRANSLOCATION EMBO JOURNAL Cory, S., Adams, J. M., GERONDAKIS, S. D., MILLER, J. F., Gamble, J., Wiener, F., Spira, J., FRANCKE, U. 1983; 2 (2): 213-216

    Abstract

    Murine plasmacytomas frequently exhibit a translocation of the distal region of chromosome 15 to the end of chromosome 12, where the immunoglobulin heavy chain locus resides. A candidate for the DNA across the chromosome fusion point is a cloned region of non-immunoglobulin DNA which in most plasmacytomas has recombined near the alpha heavy chain constant region gene. That the incoming DNA, provisionally designated LyR (lymphoid rearranging) DNA, does derive from chromosome 15 is shown here by blot analysis of DNA from two panels of somatic cell hybrids: hybridomas between an AKR T-lymphoma (Tikaut) and CBA mouse cells with a cytogenetically distinctive chromosome 15, and between mouse and Chinese hamster cells. LyR DNA segregated with chromosome 15 in all lines and the results assign LyR to the distal two thirds of that chromosome. This assignment, together with the previously reported high frequency of recombination between LyR and C(alpha) in plasmacytomas and associated alteration of LyR transcription suggests that translocation activates a LyR gene involved in plasmacytoma oncogenesis. Moreover, LyR rearrangement in certain T-lymphomas, such as the Tikaut line examined here, also implicate that gene in oncogenesis of some T-lymphomas.

    View details for Web of Science ID A1983QE55000010

    View details for PubMedID 11894928

  • ACTIVATION OF HUMAN ALPHA-1-ANTITRYPSIN GENE IN RAT HEPATOMA X HUMAN-FETAL LIVER-CELL HYBRIDS DEPENDS ON PRESENCE OF HUMAN CHROMOSOME-14 SOMATIC CELL GENETICS Pearson, S. J., TETRI, P., George, D. L., FRANCKE, U. 1983; 9 (5): 567-592

    Abstract

    In order to study the involvement of human chromosomes in the expression of liver-specific functions, we have produced somatic cell hybrids between a rat hepatoma (7777) cell line and human diploid skin fibroblasts (series XIX) or human fetal liver cells (series XXII). Production of human serum proteins was detected by immunoelectrophoretic analyses of concentrated serum-free hybrid culture supernatants. Human alpha 1-antitrypsin (AAT) was secreted by a subset of hybrids but not by the parental cells. The activated human AAT phenotype segregated concordantly with human chromosome 14 in 18 primarily HAT-selected and five azaguanine back-selected series XXII hybrids. All other chromosomes were excluded as playing a role in AAT expression. Therefore, the AAT gene (PI) is assigned to chromosome 14. This quasi-constitutive expression of a liver-specific function was not observed for the other serum proteins studied, nor was it seen in the skin fibroblast-derived hybrids (series XIX) although AAT was produced by some of them.

    View details for Web of Science ID A1983RK63300004

    View details for PubMedID 6604949

  • 2 MAIN GROUPS OF MOUSE MAJOR URINARY PROTEIN GENES, BOTH LARGELY LOCATED ON CHROMOSOME-4 EMBO JOURNAL Bishop, J. O., Clark, A. J., Clissold, P. M., HAINEY, S., FRANCKE, U. 1982; 1 (5): 615-620

    Abstract

    Fourteen different major urinary protein (MUP) genomic clones from BALB/c mice were isolated. By restriction site mapping, six of these form two sets of three overlapping clones. By the criterion of cross-hybridization, the 10 different genes fall into two groups of four (Group 1) and three (Group 2) genes, while three genes fall into neither group. Southern blot analysis of genomic DNA with Group 1 and Group 2 plasmid subclones shows that the haploid mouse (BALB/c) genome contains approximately 15 Group 1 genes, 12 Group 2 genes and at least seven MUP genes that belong to neither group. An analysis of mouse-Chinese hamster hybrid cell lines shows that most, if not all, Group 1 and Group 2 genes are located on mouse chromosome 4.

    View details for Web of Science ID A1982NZ78300014

    View details for PubMedID 6329695

  • DNA PROBES - DEVELOPMENT OF LIBRARIES ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY KUNKEL, L. M., Bruns, G., Williamson, R., Paterson, B. M., White, R., LATT, S. A., Gusella, J. F., Hauschka, S. D., Siniscalco, M., Schwartz, R. J., Housman, D., FRANCKE, U., Kedes, L. H., Hecht, B. K. 1982; 154: 53-78
  • PREVALENCE AND HERITABILITY OF DUCHENNE MUSCULAR-DYSTROPHY ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY Conneally, P. M., Brooke, M. H., Williamson, R., Hecht, B. K., Siniscalco, M., FRANCKE, U., Wolf, S., Housman, D., Nussbaum, R. L., LATT, S. A., Moss, M. L., Epstein, H. F., Boyer, S. H., CASKEY, C. T., Schwartz, R. J., Hauschka, S. D., Paterson, B. M., Strohman, R. C., NADALGINARD, B. 1982; 154: 1-24
  • SPECIAL PROBLEMS OF POLYMORPHISMS ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY White, R., LATT, S. A., Davidson, N., Housman, D., Kedes, L. H., Conneally, P. M., Gusella, J. F., Epstein, H. F., Boyer, S. H., NADALGINARD, B., Williamson, R., Nussbaum, R. L., FRANCKE, U. 1982; 154: 139-164
  • CLONING FOR AN UNKNOWN GENE ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY Davidson, N., Gusella, J. F., Paterson, B. M., NADALGINARD, B., Hauschka, S. D., Housman, D., Epstein, H. F., Brooke, M. H., Strohman, R. C., Wolf, S., Martonosi, A. N., LATT, S. A., White, R., Hecht, B. K., FRANCKE, U., Siniscalco, M., Williamson, R. 1982; 154: 25-52
  • MAPPING THE X-CHROMOSOME ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY FRANCKE, U., Epstein, H. F., Hecht, B. K., Shapiro, L. J., Siniscalco, M., Williamson, R., CASKEY, C. T., LATT, S. A., Hauschka, S. D., Boyer, S. H., Gusella, J., Schwartz, R. J., Davidson, N., KUNKEL, L. M., NADALGINARD, B., CASKEY, C. T. 1982; 154: 79-137
  • COMPARATIVE GENE-MAPPING - MURINE LAMBDA LIGHT CHAIN GENES ARE LOCATED IN REGION CEN-]B5 OF MOUSE CHROMOSOME-16 NOT HOMOLOGOUS TO HUMAN CHROMOSOME-21 CYTOGENETICS AND CELL GENETICS FRANCKE, U., DEMARTINVILLE, B., DEUSTACHIO, P., Ruddle, F. H. 1982; 33 (3): 267-271

    Abstract

    We have mapped the genes for murine immunoglobulin lambda light chains to the region of chromosome 16 proximal to band B5 by hybridizing a cDNA probe for gamma light chains to the DNA of a series of hybrid clones made between mouse fibroblasts carrying Searle's translocation, T(X;16)16H, and Chinese hamster cells. Based on homology, we predict that the human Ig gamma gene (IGL) will map to the proximal two-thirds of HSA 22.

    View details for Web of Science ID A1982PH37100012

    View details for PubMedID 6813029

  • Mapping of polypeptide genes by two-dimensional gel electrophoresis of hybrid cell extracts. Birth defects original article series Taggart, R. T., FRANCKE, U. 1982; 18 (2): 99-110

    Abstract

    A series of 18 Chinese hamster X human somatic cell hybrid subclones segregating human chromosomes were analyzed by two-dimensional gel electrophoresis to determine the chromosomal location of the human genes responsible for the expression of specific proteins. Two independent methods, side by side comparison and double label autoradiography-fluorography, were used to identify the human proteins that were unambiguously resolved from those of the Chinese hamster (CH). Of the 83 human spots resolved, 22 were determined to be associated with nine specific chromosomes or chromosome regions by correlation with quantitative cytogenetic analysis of the hybrid clones. An additional five spots were associated with three different chromosomes that had identical patterns of segregation. A total of 12 resolvable polypeptides of apparently human origin were present in all 18 hybrid subclones and may be coded for by one of the nine human autosomes that were also consistently present. Future investigations using a variety of available methods to determine the identity of the polypeptide spots will permit the mapping of genes for specific enzymes and for other classes of proteins that are not detectable by the commonly used zymogram methods.

    View details for PubMedID 7159713

  • Report of the committee on human gene mapping by recombinant DNA techniques. Birth defects original article series Skolnick, M. H., FRANCKE, U. 1982; 18 (2): 194-204

    View details for PubMedID 6760920

  • ASSIGNMENT OF THE 1ST RANDOM RESTRICTION FRAGMENT LENGTH POLYMORPHISM (RFLP) LOCUS (D14S1) TO A REGION OF HUMAN CHROMOSOME-14 AMERICAN JOURNAL OF HUMAN GENETICS DEMARTINVILLE, B., WYMAN, A. R., White, R., FRANCKE, U. 1982; 34 (2): 216-226

    Abstract

    A locus responsible for a restriction fragment length polymorphism (RFLP) has been identified by hybridization of Eco RI fragments to the random human DNA sequence in recombinant plasmid pAW101. We have examined DNA extracted from 20 human X Chinese hamster somatic cell hybrids for the presence of sequences homologous to the human insert in pAW101. The hybrids were derived from six different human donors, five of whom were heterozygous, producing two bands on Southern transfers. The presence of homologous sequences in the hybrids correlated exclusively with the presence of human chromosome 14. Three hybrids contained chromosome 14 in a frequency of greater than one per cell and were positive for two alleles. Two hybrids contained only the distal half of the long arm of 14 as part of a translocation and were still positive. These results assign the first highly polymorphic random RFPL locus (D14S1) to region q21 leads to qter of chromosome 14.

    View details for Web of Science ID A1982NH73200005

    View details for PubMedID 6280496

  • SINGLE-COPY SEQUENCE HYBRIDIZES TO POLYMORPHIC AND HOMOLOGOUS LOCI ON HUMAN X-CHROMOSOME AND Y-CHROMOSOME PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA-BIOLOGICAL SCIENCES Page, D., DEMARTINVILLE, B., Barker, D., Wyman, A., White, R., FRANCKE, U., Botstein, D. 1982; 79 (17): 5352-5356

    Abstract

    Use of a 4.5-kilobase-pair (kb) segment of single-copy DNA from a human genomic library as a hybridization probe of genomic human DNAs revealed allelic Taq I restriction fragments 10.6, 11.8, and 14.6 kb long. Among 12 unrelated individuals, all 6 males exhibited the 14.6-kb fragment in addition to one of the other fragments. Three of the females displayed 10.6- and 11.8-kb fragments, and the other three displayed only one fragment length; none had the 14.6-kb fragment. Hybridization of this probe to Taq I-digested DNAs from human-rodent hybrid cell lines (which have partial complements of human chromosomes) demonstrated segregation of the 14.6-kb fragment with the human Y chromosome and segregation of the 10.6- and 11.8-kb fragments with the human X chromosome. Furthermore, hybridization of this probe to Taq I-digested DNAs from 48 members of a single kindred revealed Y-linked inheritance of the 14.6-kb fragment and X-linked inheritance of the 10.6- and 11.8-kb fragments. These experiments demonstrate homology between single-copy sequences on the human X and Y chromosomes.

    View details for Web of Science ID A1982PE86500050

    View details for PubMedID 6291041

  • MAPPING OF POLYPEPTIDE GENES BY TWO-DIMENSIONAL GEL-ELECTROPHORESIS OF HYBRID CELL-EXTRACTS CYTOGENETICS AND CELL GENETICS Taggart, R. T., FRANCKE, U. 1982; 32 (1-4): 99-110

    Abstract

    A series of 18 Chinese hamster X human somatic cell hybrid subclones segregating human chromosomes were analyzed by two-dimensional gel electrophoresis to determine the chromosomal location of the human genes responsible for the expression of specific proteins. Two independent methods, side by side comparison and double label autoradiography-fluorography, were used to identify the human proteins that were unambiguously resolved from those of the Chinese hamster (CH). Of the 83 human spots resolved, 22 were determined to be associated with nine specific chromosomes or chromosome regions by correlation with quantitative cytogenetic analysis of the hybrid clones. An additional five spots were associated with three different chromosomes that had identical patterns of segregation. A total of 12 resolvable polypeptides of apparently human origin were present in all 18 hybrid subclones and may be coded for by one of the nine human autosomes that were also consistently present. Future investigations using a variety of available methods to determine the identity of the polypeptide spots will permit the mapping of genes for specific enzymes and for other classes of proteins that are not detectable by the commonly used zymogram methods.

    View details for Web of Science ID A1982PH37300009

    View details for PubMedID 7140373

  • ASSIGNMENT OF THE HUMAN-GENE FOR MUSCLE-TYPE PHOSPHOFRUCTOKINASE (PFKM) TO CHROMOSOME-1 (REGION CEN-]Q32) USING SOMATIC-CELL HYBRIDS AND MONOCLONAL ANTI-M ANTIBODY SOMATIC CELL GENETICS Vora, S., Durham, S., DEMARTINVILLE, B., George, D. L., FRANCKE, U. 1982; 8 (1): 95-104

    Abstract

    Human phosphofructokinase (PFK; EC 2.7.1.11) is under the control of three structural loci which encode muscle-type (M), live-type (L), and platelet-type (P) subunits; human diploid fibroblasts and leukocytes express all three loci. In order to assign human PFKM locus to a specific chromosome we have analyzed human x Chinese hamster somatic cell hybrids for the expression of human M subunits, using an anti-human M subunit-specific mouse monoclonal antibody. In 18 of 19 hybrids studied, the expression of the PFKM locus segregated concordantly with the presence of chromosome 1 (discordance rate 0.05) as indicated by chromosome and isozyme marker analysis. The discordance rates for all the other chromosomes were 0.32 or greater, indicating that the PFKM locus is on chromosome 1. For the regional mapping of PFKM, eight hybrids were studied that contained one of five distinct regions of chromosome 1. These results further localize the human PFKM locus to region cen leads to q32 chromosome 1.

    View details for Web of Science ID A1982NK73800008

    View details for PubMedID 6213050

  • REPORT OF THE COMMITTEE ON HUMAN-GENE MAPPING BY RECOMBINANT DNA TECHNIQUES CYTOGENETICS AND CELL GENETICS Skolnick, M. H., FRANCKE, U. 1982; 32 (1-4): 194-204

    View details for Web of Science ID A1982PH37300018

    View details for PubMedID 7140362

  • H-Y-ANTIGEN EXPRESSION IN PATIENTS WITH X-AUTOSOMAL TRANSLOCATIONS AND GONADAL-DYSGENESIS AMERICAN JOURNAL OF MEDICAL GENETICS Haseltine, F. P., Lynch, V. A., VanDyke, D. L., BREG, W. R., FRANCKE, U. 1982; 13 (2): 115-123

    Abstract

    Cells from three patients with early gonadal failure and a balanced reciprocal translocation involving the long arm of the X chromosome and an autosome were studied. Fibroblasts from a patient with a similar balanced reciprocal translocation but normal reproductive capabilities were also studied. Two of the four patients were found to have serologically detectable H-Y antigen on their cells. Since H-Y antigen has been found on the cells of other patients with X chromosome abnormalities but without a Y chromosome, it is thought that the X chromosome plays a role in the regulation of H-Y antigen expression. This study suggests that the long arm of the X chromosome may be involved but the location of a regulatory gene cannot be identified in these studies. These cases do not permit us to implicate H-Y antigen as a cause of gonadal dysgenesis and early gonadal failure in females who have structurally abnormal X chromosomes.

    View details for Web of Science ID A1982PH21700002

    View details for PubMedID 7137226

  • THE HUMAN GENES FOR S-ADENOSYLHOMOCYSTEINE HYDROLASE AND ADENOSINE-DEAMINASE ARE SYNTENIC ON CHROMOSOME-20 SCIENCE Hershfield, M. S., FRANCKE, U. 1982; 216 (4547): 739-742

    Abstract

    Human-Chinese hamster cell hybrids and a monoclonal antibody to human S-adenosylhomocysteine hydrolase were used to identify chromosome 20 as the location of the human gene for this enzyme. The gene for adenosine deaminase had previously been mapped to this chromosome. The activity of S-adenosylhomocysteine hydrolase is dependent in vivo on that of adenosine deaminase, since the substrates for the deaminase, adenosine and deoxyadenosine, respectively, inhibit and inactivate S-adenosylhomocysteine hydrolase in genetic or drug-induced adenosine deaminase deficiency. This functional dependence and the likelihood that S-adenosylhomocysteine hydrolase, a eukaryotic enzyme, arose later than adenosine deaminase, which occurs in prokaryotes as well as eukaryotes, suggest that the occurrence of their genes on the same chromosome may have evolutionary significance. In addition, the unusual capacity of S-adenosylhomocysteine hydrolase to form stable complexes with adenosine and its cofactor, nicotinamide adenine dinucleotide, suggest that evolution of its gene may have involved recombination of a portion of the adenosine deaminase gene with an adenine nucleotide domain-coding sequence of another preexisting gene.

    View details for Web of Science ID A1982NN26200030

    View details for PubMedID 7079734

  • ASSIGNMENT OF THE HUMAN-GENE FOR LIVER-TYPE 6-PHOSPHOFRUCTOKINASE ISOENZYME (PFKL) TO CHROMOSOME 21 BY USING SOMATIC-CELL HYBRIDS AND MONOCLONAL ANTI-L ANTIBODY PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA-BIOLOGICAL SCIENCES Vora, S., FRANCKE, U. 1981; 78 (6): 3738-3742

    Abstract

    Human 6-phosphofructokinase (PFK; ATP:D-fructose-6-phosphate 1-phosphotransferase, EC 2.7.1.11) is under the control of structural loci that code for muscle (M), liver (L), and platelet (P) subunits, which are variably expressed in different tissues; human diploid fibroblasts and leukocytes express all three genes. Random tetramerization of these subunits produces various isozymes, which can be distinguished from one another by ion exchange chromatography or by subunit-specific monoclonal antibodies. We have examined 17 somatic cell hybrids established between Chinese hamster cells and human diploid fibroblasts or leukocytes for the expression of L-type subunits of human PFK. As electrophoresis does not distinguish between Chinese hamster PFKs and human PFKs, we used an anti-human L-subunit-specific monoclonal antibody, which does not react with chinese hamster PFKs. The expression of human L subunits in the hybrids was detected by the enzyme-immunoprecipitation technique using staphylococci bearing protein A as an immunoadsorbent. Twelve out of 17 hybrids expressed human L subunits and retained chromosome 21, as determined by chromosome and isozyme marker analysis, whereas 5 did not express human PFKL and lacked chromosome 21. The mean erythrocyte PFK of seven individuals with trisomy 21 was found to be elevated (147% of normal). A specific increase in L subunits in trisomic erythrocytes was evident chromatographically by a striking increase in L4 species (50%; normal 10%) and immunologically by decreased precipitation with anti-M monoclonal antibody (50%; normal 80%). We conclude from these data that PFKL is located on chromosome 21 and that the previously noted elevation of erythrocyte PFK activity in individuals with trisomy 21 is due to a gene-dosage effect.

    View details for Web of Science ID A1981LW77700089

    View details for PubMedID 6455664

  • KARYOTYPIC ANALYSIS OF METHOTREXATE-RESISTANT AND SENSITIVE MOUSE L5178Y CELLS CYTOGENETICS AND CELL GENETICS Berenson, R. J., FRANCKE, U., Dolnick, B. J., Bertino, J. R. 1981; 29 (3): 143-152
  • GENE FOR MONOAMINE-OXIDASE TYPE-A ASSIGNED TO THE HUMAN X-CHROMOSOME JOURNAL OF NEUROSCIENCE Pintar, J. E., BARBOSA, J., FRANCKE, U., Castiglione, C. M., Hawkins, M., Breakefield, X. O. 1981; 1 (2): 166-175

    Abstract

    Inherited variations in monoamine oxidase (MAO) activity are thought to affect human behavior and expression of disease. The present study has established the chromosomal location of one of the structural genes coding for this enzyme. Mapping was carried out by somatic cell hybridization between normal human skin fibroblasts and mouse neuroblastoma cells. Selective media for growth of cells with or without hypoxanthine phosphoribosyltransferase (HPRT) activity were used to obtain hybrid lines which had retained or lost the human X chromosome, respectively. Cytogenetic techniques, isozyme analysis, and limited proteolysis and peptide mapping of [3H]pargyline-labeled MAO were used to characterize hybrid lines. With one exception, only lines containing the human X chromosome and human forms of two X-linked enzymes (phosphoglycerate kinase and glucose-6-phosphate dehydrogenase) expressed the human form of the flavin polypeptide of type A MAO. The exceptional hybrid line contained a putative translocation of part of the human X chromosome, since it expressed human forms of both MAO and phosphoglycerate kinase but neither the human form of glucose-6-phosphate dehydrogenase nor HPRT activity. This evidence indicates that the structural gene for the flavin polypeptide of MAO-A is on the human X chromosome. This represents the first chromosomal assignment of a human gene coding for an enzyme of neurotransmitter metabolism. This information will help to elucidate the structure of MAO and modes of its inheritance in the human population.

    View details for Web of Science ID A1981MP44700006

    View details for PubMedID 7196439

  • CONSERVED AUTOSOMAL SYNTENIC GROUP ON MOUSE (MMU) CHROMOSOME 15 AND HUMAN (HSA) CHROMOSOME-22 - ASSIGNMENT OF A GENE FOR ARYLSULFATASE-A TO MMU 15 AND REGIONAL MAPPING OF DIA1, ARSA, AND ACO2 ON HSA 22 CYTOGENETICS AND CELL GENETICS FRANCKE, U., TETRI, P., Taggart, R. T., OLIVER, N. 1981; 31 (2): 58-69

    Abstract

    We have utilized a panel of Chinese hamster x mouse somatic cell hybrids segregating mouse chromosomes to assign a gene for arylsulfatase A (ARSA) to mouse chromosome 15. Considering our previous assignment of a gene for diaphorase-1 (DIA1) to the same mouse chromosome, we have evidence for another syntenic relationship that has been conserved, since the homologous loci for human ARSA and DIA1 are both located on human chromosome 22. Because MMU 15 and HSA 22 are quite dissimilar in size and banding patterns, we have attempted to identify the conserved portion by regional mapping of human DIA1 and ARSA using somatic cell hybrids segregating a human chromosome translocation t(15;22)(q14;q13.31). The results assign human DIA1 and ARSA to the distal sub-band of 22q13 (region 22q13.31 leads to qter). The locus for mitochondrial aconitase (ACO2) has been separated by the breakpoint from DIA1 and ARSA and is located more proximally.

    View details for Web of Science ID A1981MT29100004

    View details for PubMedID 6118238

  • ABSENCE OF H-Y-ANTIGEN IN AN XY FEMALE WITH CAMPOMELIC DYSPLASIA HUMAN GENETICS Puck, S. M., Haseltine, F. P., FRANCKE, U. 1981; 57 (1): 23-27

    Abstract

    We have studied a stillborn infant who had the clinical and radiographic characteristics of campomelic dysplasia. External and internal genitalia were those of a normal female, except for slight enlargement of the clitoris. Microscopic examination of the ovaries revealed some areas resembling immature dysgenetic testicular tissue. Karyotypes from lymphocyte and fibroblast cultures were 46,XY with a structurally normal Y chromosome and no evidence of mosaicism. H-Y antigen was not detected on the fibroblasts in repeated assays using Raji cells as target cells after absorption. The "sex- reversal" (chromosomal male Leads to phenotypic female) previously noted in patients with autosomal recessive campomelic dysplasia thus may be mediated through lack of detectable H-Y antigen on the cell surface. It appears that the mutation leading to campomelic dysplasia interferes with normal H-Y antigen expression.

    View details for Web of Science ID A1981LJ61100003

    View details for PubMedID 7196381

  • HIGH-RESOLUTION IDEOGRAMS OF TRYPSIN-GIEMSA BANDED HUMAN-CHROMOSOMES CYTOGENETICS AND CELL GENETICS FRANCKE, U. 1981; 31 (1): 24-32

    View details for Web of Science ID A1981MP73100002

    View details for PubMedID 7297127

  • ASSIGNMENT OF GENES TO THE HUMAN X-CHROMOSOME BY THE TWO-DIMENSIONAL ELECTROPHORETIC ANALYSIS OF TOTAL CELL-PROTEINS FROM RODENT-HUMAN SOMATIC-CELL HYBRIDS AMERICAN JOURNAL OF HUMAN GENETICS Cox, D. R., FRANCKE, U., Epstein, C. J. 1981; 33 (4): 495-512

    Abstract

    The technique of two-dimensional (2-D) gel electrophoresis was sued to identify five human X-linked gene products in crude cell extracts of mouse-human and Chinese hamster-human somatic cell hybrids. The human origin of these five polypeptides was demonstrated by their comigration with human fibroblast proteins and their failure to comigrate with polypeptides in extracts from the mouse or hamster parental cells. All five polypeptides were present in extracts of rodent-human hybrids that contained a human X chromosome, but were not found in extracts of cells that lacked a human X chromosome. Chromosome analysis of the hybrid clones revealed that the human X chromosome is both necessary and sufficient for the expression of the five polypeptides, designated pX-24, pX-27, pX-37, pX-40, and pX-56. pX-56 can be identified as the human X-linked enzyme glucose-6-phosphate dehydrogenase (G6PD) (E.C.1.1.1.49), while polypeptides pX-24, pX-27, pX-37 and pX-40 have molecular properties unlike those of known human X-linked gene products. pX-24 appears to be a membrane-bound protein that maps to the distal portion of the long arm of the human X chromosome, while pX-27, pX-37, and pX-40 are soluble proteins that map to the proximal long arm or to the short arm of the human X chromosome. 2-D gel electrophoretic analysis of extracts from somatic cell hybrids provides a general method for identifying polypeptides in crude cell extracts coded for by any specific chromosome and can be used to study primary gene products not previously amenable to genetic analysis.

    View details for Web of Science ID A1981LZ34800001

    View details for PubMedID 7196152

  • REQUIREMENT OF THE HUMAN CHROMOSOME-11 LONG ARM FOR REPLICATION OF HERPES-SIMPLEX VIRUS TYPE-1 IN NON-PERMISSIVE CHINESE-HAMSTER X HUMAN-DIPLOID FIBROBLAST HYBRIDS SOMATIC CELL GENETICS FRANCKE, U., Francke, B. 1981; 7 (2): 171-191

    Abstract

    Somatic cell hybrids between Chinese hamster (CH) lung cells (V79/380-6), nonpermissive for productive infection by herpes simplex type 1 (HSV-1), and permissive human diploid cells support productive HSV-1 infection as long as they retain human chromosome 11. Human chromosome 3 has been reported to complement nonpermissivity in (CH) Don cells (1). Intraspecies hybrids between Don/a3 and V79/380-6 cells, however, did not support HSV-1 replication, indicating lack of complementation. The block in both nonpermissive CH cell lines was determined to involve a step beyond replication of the parental viral DNA. In cell hybrids between nonpermissive Don/a23 cells and human fibroblasts containing a t(11;15) (p11;p12) translocation, HSV-1 production was dependent solely on the presence of either human chromosome 11 or the der(11) (p11 leads to qter) translocation product containing the long arm of chromosome 11. Chromosome 3 was excluded by a discordancy rate of 59%. We conclude that the long arm of human chromosome 11 carries one or more genes coding for host functions necessary for the production of progeny HSV-1 DNA.

    View details for Web of Science ID A1981LH17400005

    View details for PubMedID 6270835

  • THE GENES FOR GROWTH-HORMONE AND CHORIONIC SOMATOMAMMOTROPIN ARE ON THE LONG ARM OF HUMAN CHROMOSOME-17 IN REGION Q21-]QTER HUMAN GENETICS George, D. L., Phillips, J. A., FRANCKE, U., Seeburg, P. H. 1981; 57 (2): 138-141

    Abstract

    We used a cloned cDNA probe for human growth hormone and Southern blotting techniques to analyze DNA from a series of rodent x human somatic cell hybrids for the presence of growth hormone-related sequences. Our results provide evidence for the assignment of the genes for growth hormone and chorionic somatomammotropin as well as a growth hormone-like gene to human chromosome 17. Analysis of mouse x human hybrid cells containing only part of the long arm of chromosome 17 enabled us to localize these genes to region 17q21 to 17qter.

    View details for Web of Science ID A1981LN11200004

    View details for PubMedID 6262212

  • Karyotypic analysis of methotrexate-resistant and sensitive mouse L5178Y cells. Cytogenetics and cell genetics Berenson, R. J., FRANCKE, U., Dolnick, B. J., Bertino, J. R. 1981; 29 (3): 145-152

    View details for PubMedID 6939521

  • Cytogenetic approaches to mouse models of human genetic diseases. American journal of pathology FRANCKE, U. 1980; 101 (3): S41-51

    View details for PubMedID 7192937

  • CHROMOSOME ASSIGNMENT OF A MURINE GLUCOCORTICOID RECEPTOR GENE (GRL-1) USING INTRASPECIES SOMATIC-CELL HYBRIDS CELL FRANCKE, U., Gehring, U. 1980; 22 (3): 657-664

    View details for Web of Science ID A1980KW22300006

    View details for PubMedID 7460010

  • Report of the committee on the genetic constitution of chromosome 6. Birth defects original article series FRANCKE, U., Weitkamp, L. R. 1980; 15 (11): 32-38

    View details for PubMedID 6938253

  • HOMOGENEOUSLY STAINING CHROMOSOME REGIONS AND DOUBLE MINUTES IN A MOUSE ADRENOCORTICAL TUMOR-CELL LINE CYTOGENETICS AND CELL GENETICS George, D. L., FRANCKE, U. 1980; 28 (4): 217-226

    Abstract

    We have used a number of banding techniques to analyze the chromosomal content of two sublines of the steroid-secreting mouse adrenocortical tumor cell line Y1. One of the sublines contains marker chromosomes with large "homogeneously staining regions" (HSRs). The other subline contains numerous double minutes, but no HSR. The presence of marker chromosomes shared by both sublines indicates their derivation from a common precursor. Both sublines are karyotypically stable, have similar growth rates, and exhibit positive staining for delta 5, 3 beta-hydroxysteroid dehydrogenase.

    View details for Web of Science ID A1980KU60600001

    View details for PubMedID 6936133

  • SUPERMELANOTIC HYBRIDS DERIVED FROM MOUSE MELANOMAS AND NORMAL MOUSE CELLS SOMATIC CELL GENETICS Halaban, R., Nordlund, J., FRANCKE, U., MOELLMANN, G., Eisenstadt, J. M. 1980; 6 (1): 29-44

    Abstract

    Hybrids formed between HPRT- Cloudman mouse melanoma and normal cells were isolated. The parental origin of the hybrids was verified by isoenzyme and karyotype analyses. These hybrid cells differed in two major characteristics from hybrids of melanoma and established fibroblastic cells. (1) They grew as tumors when injected into mice, and (2) they expressed differentiated melanocytic functions. At least one of the differentiated functions was overexpressed. The specific activity of tyrosinase was 3-20 times higher in the hybrid cells than in the parental mouse melanoma. The overexpression of tyrosinase in these hybrid cells has been stable for more than a year, has been transmitted to subclones of the original hybrid cell lines, and has been expressed in tumors that grew after injections of hybrid cells into animals.

    View details for Web of Science ID A1980JJ89400003

    View details for PubMedID 6768142

  • THE ANIRIDIA-WILMS-TUMOR-ASSOCIATION - THE CRITICAL ROLE OF CHROMOSOME BAND 11P13 CANCER GENETICS AND CYTOGENETICS Riccardi, V. M., HITTNER, H. M., FRANCKE, U., Yunis, J. J., Ledbetter, D., Borges, W. 1980; 2 (2): 131-137
  • A METHOTREXATE-RESISTANT SUBLINE OF MOUSE L1210 LEUKEMIA-CELLS CONTAINING HIGH-LEVELS OF DIHYDROFOLATE-REDUCTASE AND WITH A HOMOGENEOUSLY STAINING REGION ON CHROMOSOME-4 ONCODEVELOPMENTAL BIOLOGY AND MEDICINE Trowsdale, J., Hoch, J. A., FRANCKE, U. 1980; 1 (6): 369-374
  • DUPLICATION 12Q MOSAICISM IN 2 UNRELATED PATIENTS WITH A SIMILAR SYNDROME AMERICAN JOURNAL OF MEDICAL GENETICS HARROD, M. J., Byrne, J. B., Dev, V. G., FRANCKE, U. 1980; 7 (2): 123-129

    Abstract

    Two unrelated children with a similar syndrome were found to have mosaicism for a cell line containing one chromosome 12 with an additional faintly G-banding staining region that apparently represents a duplication of the distal portion of the long arm. The homology and the other chromosomes are normal, as are the parental chromosomes. The remarkable phenotypic similarity of the 2 patients and their resemblance to 2 previously reported patients with duplication of the same chromosome region suggests that duplication 12q24 results in a clinically identifiable malformation syndrome.

    View details for Web of Science ID A1980KU16600005

    View details for PubMedID 7468644

  • Variation in human HPRT and its relationship to neurologic and behavioral manifestations. Advances in experimental medicine and biology BAKAY, B., Nissinen, E., Sweetman, L., FRANCKE, U., Nyhan, W. L. 1980; 122A: 317-325

    View details for PubMedID 7424650

  • BETA-GLOBIN GENE IS ON THE SHORT ARM OF HUMAN CHROMOSOME-11 NATURE SANDERSHAIGH, L., Anderson, W. F., FRANCKE, U. 1980; 283 (5748): 683-686

    Abstract

    Investigations on the regulation of human globin gene expression are assisted by a knowledge of the chromosomal location of the globin genes. Previous studies have mapped the alpha-globin locus to human chromosome (HC) 16, and have shown that the human globin gene complex gamma-delta-beta co-segregates with lactate dehydrogenase A (LDH-A) and the presence of HC 11 in somatic cell hybrids. Radioactively labelled globin complementary DNA (cDNA) probes were used in molecular hybridisation experiments to determine the chromosomal locations of the alpha- and beta-globin genes. When human x rodent somatic cell hybrids are used which contain well-defined parts of human chromosomes, direct mapping of genes of chromosomal regions or single bands is possible. We have regionally localised the human beta-globin gene using two sets of hybrid cell lines: (1) Chinese hamster x human hybrid cells containing the HC 11 long arm or both the short and long arms and (2) mouse x human hybrids containing only the HC 11 short arm. The techniques of liquid molecular hybridisation and Southern blotting with 32P-labelled human beta-globin cDNA (from plasmid JW102) have been used to localise the beta-globin gene sequence to region 11p11 leads to 11p15. Similar results were reported recently by Jeffreys et al.

    View details for Web of Science ID A1980JE68400054

    View details for PubMedID 7354859

  • ASSIGNMENT OF THE GENE FOR NADH DIAPHORASE DIA-1 TO MOUSE CHROMOSOME-15 SOMATIC CELL GENETICS Taggart, R. T., TETRI, P., FRANCKE, U. 1980; 6 (6): 769-776

    View details for Web of Science ID A1980KT77800008

    View details for PubMedID 6893763

  • COMPARATIVE GENE-MAPPING - ORDER OF LOCI ON THE X-CHROMOSOME IS DIFFERENT IN MICE AND HUMANS PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA-BIOLOGICAL SCIENCES FRANCKE, U., Taggart, R. T. 1980; 77 (6): 3595-3599

    Abstract

    For comparative studies we have used the somatic cell hybridization approach to regionally map genes on the mouse X chromosome. Fibroblasts from a mouse with the balanced reciprocal translocation T(XD;16B5)16H were fused with a Chinese hamster cell line (V79/380-6) deficient in activity of the enzyme hypoxanthine phosphoribosyltransferase (HPRT). Interpecific cell hybrids were initially selected for retention of the mouse translocation chromosome carrying the Hprt gene. Subsequently, hybrid clones were counterselected to force segregation of this chromosome. Selected and counterselected hybrid clones were analyzed for their chromosome content by trypsin/Giemsa banding and for expression of the mouse forms of the X-linked enzymes HPRT and alpha-galactosidase (GALA) by isoelectric focusing. The results indicate that the breakpoint on the mouse X chromosome (in band XD) has separated the genes for HPRT (Hprt) and for GALA (Ags). Hprt is proximal to the breakpoint in region Xcen-XD and Ags is distal in region XD-Xter. The gene order in the mouse (centromere-Hprt-Ags) is therefore inverted when compared to the order of the homologous loci on the long arm of the human X (centromere-GALA-HPRT).

    View details for Web of Science ID A1980JY17400096

    View details for PubMedID 6251472

  • DIFFUSE POLYCLONAL B-CELL LYMPHOMA DURING PRIMARY INFECTION WITH EPSTEIN-BARR VIRUS NEW ENGLAND JOURNAL OF MEDICINE Robinson, J. E., Brown, N., Andiman, W., Halliday, K., FRANCKE, U., Robert, M. F., ANDERSSONANVRET, M., Horstmann, D., Miller, G. 1980; 302 (23): 1293-1297

    View details for Web of Science ID A1980JT86300006

    View details for PubMedID 6245364

  • THE PENTA-X SYNDROME JOURNAL OF MEDICAL GENETICS Monheit, A., FRANCKE, U., Saunders, B., LYONSJONES, K. 1980; 17 (5): 392-396

    Abstract

    A child is presented with a 49,XXXXX chromosomal constitution bringing to 12 the total number of children described with this karyotype. Comparison of this child's features with previously reported cases indicates a clinically recognisable specific pattern of malformations referred to as the penta-X syndrome. X chromosome replication studies using BrdU labelling in the patient's cells clearly showed that the four presumably inactive X chromosomes were late replicating but not in a strictly synchronous fashion.

    View details for Web of Science ID A1980KL11800014

    View details for PubMedID 7218280

  • KARYOTYPE EVOLUTION OF THE SIMIAN-VIRUS 40-TRANSFORMED HUMAN CELL LINE LNSV CYTOGENETICS AND CELL GENETICS Begovich, A., FRANCKE, U. 1979; 23 (1-2): 3-11

    Abstract

    We have used trypsin-Wright's banding ("GTG-banding") to analyze the chromosome content in two sublines of the SV40-transformed human cell line LNSV, derived from fibroblasts of a patient with HPRT deficiency. Both LNSV sublines (GM-847 and "LNSV") were heteroploid and showed considerable numerical and structural chromosome variability. Nineteen rearranged chromosomes which were observed at high frequency have been set aside as "marker chromosomes," and their probable derivation from normal human chromosomes has been described in PARIS CONFERENCE (1971) nomenclature. Heterogeneity within these uncloned sublines appears to increase with time in culture, and no evidence was found for evolution of a karyotypically stable cell population. The results are of general significance for cell genetic studies using established cell lines.

    View details for Web of Science ID A1979GC06400001

    View details for PubMedID 216530

  • LOCALIZATION OF THE G-GAMMA-GLOBIN, A-GAMMA-GLOBIN, DELTA-GLOBIN AND BETA-GLOBIN GENES ON THE SHORT ARM OF HUMAN CHROMOSOME-11 NATURE Jeffreys, A. J., Craig, I. W., FRANCKE, U. 1979; 281 (5732): 606-608

    Abstract

    Human-mouse somatic cell hybrids have proved invaluable in assigning human genes to their respective human chromosomes. To date, the success of this approach has depended on identifying human proteins which are synthesised in hybrid cells containing a small number of human chromosomes. Consequently, chromosome assignment has been limited mainly to human proteins which are expressed in man-mouse somatic cell hybrids and for which a suitable assay, usually electrophoretic or immunological, exists to distinguish between the human and murine homologous proteins. This technique is therefore unsuitable for the assignment of those human genes which are expressed only in differential cells and not in hybrid cells. Here, we describe how nucleic acid hybridisation and restriction endonuclease mapping of DNA can be combined to test for the presence of human structural gene sequences within hybrid cell DNA. This method can be used to assign any purified human DNA sequence to a human chromosome, and does not require the DNA sequence to be expressed in man-mouse hybrid cells.

    View details for Web of Science ID A1979HQ78400073

    View details for PubMedID 492329

  • SPONTANEOUS EXPRESSION OF MURINE ENDOGENOUS VIRUSES IN HAMSTER X MOUSE HYBRID-CELLS INTERNATIONAL JOURNAL OF CANCER Park, S. S., Gazdar, A. F., Lalley, P. A., Minna, J. D., FRANCKE, U. 1979; 23 (1): 52-61

    View details for Web of Science ID A1979GG02000010

    View details for PubMedID 215563

  • ANIRIDIA-WILMS TUMOR ASSOCIATION - EVIDENCE FOR SPECIFIC DELETION OF 11P13 CYTOGENETICS AND CELL GENETICS FRANCKE, U., Holmes, L. B., Atkins, L., Riccardi, V. M. 1979; 24 (3): 185-192

    Abstract

    A 7-year-old boy with aniridia, Wilms' tumor, and mental retardation, previously reported as having an interstitial deletion of the short arm of chromosome 8 resulting from a t(8p+;11q-) translocation (Ladda et al., 1974), has been restudied using high-resolution trypsin-Giemsa banding of prometaphase chromsomes. The results revealed a complex rearrangement with four break points in 8p, 11p, and 11q, leading to a net loss of an interstitial segment of 11p (region p1407 yields p1304) but not of 8p. His red blood cells contained normal activities of glutathione reductase (gene on 8p) and lactate dehydrogeanse A (gene on 11p12), indicating a gene dosage consistent with the chromosomal findings. The revised interpretation of this case agrees with seven others reported as having aniridia and interstitial 11p deletions in establishing the distal half of band 11p13 as the site of gene(s) which lead to aniridia and predispose to Wilms' tumor if present in a hemizygous state. Possible relationships between heterozygous deletion of specific chromosomal bands 11p13 and 13q14 and the autosomal dominant disorders aniridia, Wilms' tumor, and retinoblastoma, respectively, are discussed.

    View details for Web of Science ID A1979HM38800008

    View details for PubMedID 225131

  • PARTIAL TRIPLICATION AND DELETION OF 13Q - STUDY OF A FAMILY PRESENTING WITH BILATERAL RETINOBLASTOMAS CLINICAL GENETICS Riccardi, V. M., HITTNER, H. M., FRANCKE, U., PIPPIN, S., Holmquist, G. P., Kretzer, F. L., Ferrell, R. 1979; 15 (4): 332-345

    Abstract

    This report compares the pathogenetic influences of selective deletion and triplicaton of chromosome 13 derived from a familial 12;13 insertional translocation. In the proband a heritable chromosomal basis for his bilateral retinoblastomas is established [46,XY,del (13) (pter leads to q12.5: :q22.1 leads to qter)mat], and in his sister the relatively modest effects of triplication of the mid-portions of 13q are demonstrated [46,XX,ins(12;13) (12pter leads to 12p11.2: :13q22.1 leads to 13q12.5: :12p11.2 leads to 12qter)mat]. Qualitative and quantitative gene marker studies and chromosomal staining techniques to differentiate timing of DNA replication failed to indicate functional gene changes about the breakpoints.

    View details for Web of Science ID A1979GU99000007

    View details for PubMedID 436330

  • PARTIAL TRISOMY FOR THE DISTAL LONG ARM OF CHROMOSOME-5 (REGION-Q34-]QTER) - NEW CLINICALLY RECOGNIZABLE SYNDROME CLINICAL GENETICS Curry, C. J., Loughman, W. D., FRANCKE, U., Hall, B. D., Golbus, M. S., Derstine, J., Epstein, C. J. 1979; 15 (5): 454-461

    Abstract

    This report describes a family in which eight individuals in three generations had mental retardation in association with a characteristic pattern of clinical problems and physical abnormalities including short stature, eczema, hernias, delayed puberty, dysmorphic facies and digital anomalies. The family history was consistent with a chromosomal rearrangement with transmission through balanced carriers. Routine ASG banding studies showed extra chromosomal material on a chromosome 16 but failed to demonstrate any differences between the affected individuals and the presumed carriers. However, subsequent studies utilizing trypsin banding and microspectrophotometry of individual chromosomes demonstrated that the affected individuals were partially trisomic for the distal band of the long arm of chromosome 5 and that 0.273 units of a chromosome 5 were translocated to chromosome 16. This definitive cytogenetic diagnosis permitted accurate prenatal diagnosis to be carried out on the fetus of a balanced carrier female. The application of these techniques to previously obscure familial dysmorphic syndromes is recommended.

    View details for Web of Science ID A1979GY44400012

    View details for PubMedID 445868

  • ANIRIDIA CAUSED BY A HERITABLE CHROMOSOME-11 DELETION OPHTHALMOLOGY HITTNER, H. M., Riccardi, V. M., FRANCKE, U. 1979; 86 (6): 1173-1183

    Abstract

    A child with aniridia, multiple anomalies, severe failure to thrive, and severe psychomotor retardation is shown to have a syndrome similar to, though more severe than, other patients with overlapping deletions of the short arm of chromosome 11 (Pediatrics 64:604, 1978). Her deletion (46,XX,del [11p] [pter yields p14::p11.3 yields qter]) was derived from her mother, who has a chromosome 11 shift (46,XX,der [11] [pter yields p14::p11.3 yields q22::p14 yields p11.3::q22 yields qter]). The significance of del (11p) in the aniridia-Wilms' tumor association is discussed, and the del (11p) basis for aniridia is compared with other genetic bases for aniridia.

    View details for Web of Science ID A1979JF09300024

    View details for PubMedID 230439

  • REQUIREMENT OF HUMAN-CHROMOSOMES 19, 6 AND POSSIBLY 3 FOR INFECTION OF HAMSTER X HUMAN HYBRID-CELLS WITH BABOON M7 TYPE-C VIRUS CELL Brown, S., OIE, H. K., Gazdar, A. F., Minna, J. D., FRANCKE, U. 1979; 18 (1): 135-143

    View details for Web of Science ID A1979HL27000015

    View details for PubMedID 228859

  • UTILIZATION OF PURINES BY AN HPRT VARIANT IN AN INTELLIGENT, NONMUTILATIVE PATIENT WITH FEATURES OF THE LESCH-NYHAN SYNDROME PEDIATRIC RESEARCH BAKAY, B., Nissinen, E., Sweetman, L., FRANCKE, U., Nyhan, W. L. 1979; 13 (12): 1365-1370

    Abstract

    The patient, H.Chr.B., was among the first reported with hyperuricemia and central nervous system symptoms. He has been found to have a variant of hypoxanthine guanine phosphoribosyl transferase (HPRT; E.C.2.4.2.8) distinct from the enzyme present in patients with the Lesch-Nyhan syndrome. The patient had chroeoathetosis, spasticity, dysarthric speech, and hyperuricemia. However, his intelligence was normal and he had no evidence of self-mutilation. There was no activity of HPRT in the lysates of erythrocytes and cultured fibroblasts when analyzed in the usual manner. Using a newly developed method for the study of purine metabolism in intact cultured cells, this patient was found to metabolize some 9% of 8-14C-hypoxanthine, and 90% of the isotope utilized was converted to adenine and guanine nucleotides. In contrast, cells from patients with the Lesch-Nyhan syndrome were virtually completely unable to convert hypoxanthine to nucleotides. The patient's fibroblasts were even more efficient in the metabolism of 8-14C-guanine, which was utilized to the extent of 27%, over 80% of which was converted to guanine and adenine nucleotides. The growth of the cultured fibroblasts of this patient was intermediate in media containing hypoxanthine aminopterin thymidine (HAT), whereas the growth of Lesch-Nyhan cells was inhibited and normal cells grew normally. Similarly in 8-azaguanine, 6-thioguanine, and 8-azahypoxanthine, the growth of the patient's cells was intermediate between normal and Lesch-Nyhan cells. These observations provide further evidence for genetic heterogeneity among patients with disorders in purine metabolism involving the HPRT gene. They document that this famous patient did not have the Lesch-Nyhan syndrome.

    View details for Web of Science ID A1979HY57300013

    View details for PubMedID 523196

  • REPORT OF THE COMMITTEE ON THE GENETIC CONSTITUTION OF CHROMOSOME-6 CYTOGENETICS AND CELL GENETICS FRANCKE, U., Weitkamp, L. R. 1979; 25 (1-4): 32-38

    View details for Web of Science ID A1979JN85700005

    View details for PubMedID 396126

  • ASSIGNMENT OF THE GENE FOR CYTOPLASMIC SUPEROXIDE-DISMUTASE (SOD-1) TO A REGION OF CHROMOSOME-16 AND OF HPRT TO A REGION OF THE X-CHROMOSOME IN THE MOUSE PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA FRANCKE, U., Taggart, R. T. 1979; 76 (10): 5230-5233

    Abstract

    In the search for homologous chromosome regions in man and mouse, the locus for cytoplasmic superoxide dismutase (SOD-1; superoxide:superoxide oxidoreductase, EC 1.15.1.1) is of particular interest. In man, the SOD-1 gene occupies the same subregion of chromosome 21 that causes Down syndrome when present in triplicate. Although not obviously implicated in the pathogenesis, SOD-1 is considered to be a biochemical marker for this aneuploid condition. Using a set of 29 mouse-Chinese hamster somatic cell hybrids, we assign Sod-1 to mouse chromosome 16. Isoelectric focusing permits distinction between mouse and Chinese hamster isozymes, and trypsin/Giemsa banding distinguishes mouse from Chinese hamster chromosomes. The mouse fibroblasts used were derived from a male mouse carrying Searle's T(X;16)16H reciprocal translocation in which chromosomes X and 16 have exchanged parts. Analysis of informative hybrids leads to regional assignment of Sod-1 to the distal half of mouse chromosome 16 (16B4 --> ter). Because the Chinese hamster cell line (380) used for cell hybridization is deficient in hypoxanthine phosphoribosyltransferase (HPRT; IMP: pyrophosphate phosphoribosyltransferase, EC 2.4.2.8), that part of the mouse X chromosome carrying the complementing Hprt gene can be identified by selection in hypoxanthine/aminopterin/thymidine medium and counterselection in 8-azaguanine. Mouse Hprt is on the X(T) translocation product containing the proximal region X cen --> XD.

    View details for Web of Science ID A1979HR46000102

    View details for PubMedID 291939

  • IMMEDIATE CHROMOSOME DIAGNOSIS ON BONE-MARROW CELLS - AID TO MANAGEMENT OF THE MALFORMED NEWBORN-INFANT JOURNAL OF PEDIATRICS FRANCKE, U., Brown, M. G., Jones, K. L. 1979; 94 (2): 289-292

    View details for Web of Science ID A1979GG11300029

    View details for PubMedID 762626

  • CHROMOSOME 3Q DUPLICATION AND THE BRACHMANN-DELANGE SYNDROME (BDLS) JOURNAL OF PEDIATRICS FRANCKE, U., Opitz, J. M. 1979; 95 (1): 161-162

    View details for Web of Science ID A1979HB60400052

    View details for PubMedID 480006

  • CHROMOSOMAL IMBALANCE IN ANIRIDIA-WILMS TUMOR ASSOCIATION - 11P INTERSTITIAL DELETION PEDIATRICS Riccardi, V. M., Sujansky, E., Smith, A. C., FRANCKE, U. 1978; 61 (4): 604-610

    Abstract

    The triad of aniridia, ambiguous genitalia, and mental retardation (AGR triad) is the characteristic clinical feature of three unrelated patients with previously unreported chromosome 11 short arm interstitial deletions. A Wilms' tumor in one patient establishes one cause for the aniridia-Wilms' tumor association. The genetic heterogeneity of aniridia, the AGR triad, and Wilms' tumor are demonstrated, and Wilms' tumor is indicated to be a neoplastic birth defect which can result from a variety of embryologic insults, some of which may be chromosomal or heritable.

    View details for Web of Science ID A1978EU67200021

    View details for PubMedID 208044

  • REPORT OF THE COMMITTEE ON THE GENETIC CONSTITUTION OF CHROMOSOME-6 CYTOGENETICS AND CELL GENETICS Weitkamp, L., FRANCKE, U. 1978; 22 (1-6): 92-105
  • SILVER STAINING STUDIES ON SHORT ARM VARIANT OF HUMAN CHROMOSOME 17 HUMAN GENETICS OLIVER, N., FRANCKE, U., Taylor, K. M. 1978; 42 (1): 79-82

    Abstract

    The absence of silver grain precipitation on human chromosome 17, consistently observed in four patients displaying the 17p variant, supports the hypothesis that the 17p variant is a structural heteromorphism rather than translocated satellite material.

    View details for Web of Science ID A1978FA42100013

    View details for PubMedID 77246

  • PRECISE MAPPING OF GENES FOR PHOSPHOGLUCOMUTASE-1 AND URIDINE MONOPHOSPHATE KINASE ON THE SHORT ARM OF HUMAN CHROMOSOME-1 CYTOGENETICS AND CELL GENETICS FRANCKE, U., George, D. L. 1978; 22 (1-6): 384-388

    View details for Web of Science ID A1978GU98300065

    View details for PubMedID 222544

  • REGIONAL ASSIGNMENT OF GENES FOR MANNOSE PHOSPHATE ISOMERASE, PYRUVATE KINASE-3, AND BETA-2-MICROGLOBULIN EXPRESSION ON HUMAN-CHROMOSOME 15 BY HYBRIDIZATION OF CELLS FROM A T(15,22) (Q14,Q13.3) TRANSLOCATION CARRIER CYTOGENETICS AND CELL GENETICS OLIVER, N., FRANCKE, U., Pellegrino, M. A. 1978; 22 (1-6): 506-510

    View details for Web of Science ID A1978GU98300096

    View details for PubMedID 88301

  • CONSERVATION OF AUTOSOMAL GENE SYNTENY GROUPS IN MOUSE AND MAN NATURE Lalley, P. A., Minna, J. D., FRANCKE, U. 1978; 274 (5667): 160-163

    View details for Web of Science ID A1978FF64300043

    View details for PubMedID 662012

  • COMPARATIVE GENE-MAPPING - LINKAGE RELATIONSHIPS OF THE HOMOLOGOUS GENES FOR PHOSPHOGLUCOMUTASE AND PEPTIDASE-S ARE CONSERVED IN MAN AND MOUSE CYTOGENETICS AND CELL GENETICS Lalley, P. A., FRANCKE, U., Minna, J. D. 1978; 22 (1-6): 573-576

    View details for Web of Science ID A1978GU98300113

    View details for PubMedID 752543

  • Clinical syndromes associated with partial duplications of chromosomes 2 and 3: dup(2p),dup(2q),dup(3p),dup(3q). Birth defects original article series FRANCKE, U. 1978; 14 (6C): 191-217

    View details for PubMedID 365267

  • EVIDENCE FOR LOCALIZATION OF THE GENE FOR HEXOSAMINIDASE-B TO THE CEN-]Q13 REGION OF HUMAN CHROMOSOME-5 USING MOUSE X HUMAN HYBRID-CELLS CYTOGENETICS AND CELL GENETICS George, D. L., FRANCKE, U. 1978; 22 (1-6): 408-411

    View details for Web of Science ID A1978GU98300071

    View details for PubMedID 752513

  • REGIONAL MAPPING OF HUMAN GENES FOR PHOSPHOGLUCOMUTASE-1 ON CHROMOSOME-1 AND BETA-GLUCURONIDASE ON CHROMOSOME 7 USING MOUSE X HUMAN HYBRIDS HUMAN HEREDITY George, D. L., FRANCKE, U. 1978; 28 (3): 161-170

    Abstract

    Two independent mouse-human somatic cell hybrid clones contained different, de novo chromosome rearrangements involving the short arm of human chromosome 1. One hybrid clone contained a translocation between human chromosomes 1 and 7; the other clone contained a rearrangement product between human chromosomes 1 and 14. Analysis of these clones for expression of genes previously assigned to chromosome 7 and to the short arm of chromosome 1 provided evidence for localization of PGM--1 in segment 1p22.1 leads to 1p31.1, AK--2, ENO--1 and UMPK in region 1pter leads to 1p31.1, and GUS in region 7 pter leads to 7q22. The results have been used to examine the relationship between cytologic and genetic map distances on the short arm of chromosome 1.

    View details for Web of Science ID A1978EZ33100001

    View details for PubMedID 206510

  • ASSIGNMENT OF A GENE REQUIRED FOR INFECTION WITH ENDOGENOUS BABOON VIRUS TO HUMAN CHROMOSOME-19 CYTOGENETICS AND CELL GENETICS Brown, S., OIE, H., FRANCKE, U., Gazdar, A. F., Minna, J. D. 1978; 22 (1-6): 239-242

    View details for Web of Science ID A1978GU98300032

    View details for PubMedID 222542

  • HOMOLOGOUS GENES FOR ENOLASE, PHOSPHOGLUCONATE DEHYDROGENASE, PHOSPHOGLUCOMUTASE, AND ADENYLATE KINASE ARE SYNTENIC ON MOUSE CHROMOSOME-4 AND HUMAN CHROMOSOME-1P PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Lalley, P. A., FRANCKE, U., Minna, J. D. 1978; 75 (5): 2382-2386

    Abstract

    It is possible to generate interspecific somatic cell hybrids that preferentially segregate mouse chromosomes, thus making possible mapping of mouse genes. Therefore, comparison of the linkage relationships of homologous genes in man and mouse is now possible. Chinese hamster x mouse somatic cell hybrids segregating mouse chromosomes were tested for the expression of mouse enolase (ENO-1; EC 4.2.1.11, McKusick no. 17245), 6-phosphogluconate dehydrogenase [PGD; EC 1.1.1.44, McKusick no. 17220], phosphoglucomutase-2 (PGM-2; EC 2.7.5.1, McKusick no. 17190), and adenylate kinase-2 (AK-2; EC 2.7.4.3, McKusick no. 10302). In man, genes coding for the homologous forms of these enzymes have been assigned to the short arm of human chromosome 1. Analysis of 41 primary, independent, hybrid clones indicated that, in the mouse, ENO-1 and AK-2 are syntenic with PGD and PGM-2 and therefore can be assigned to mouse chromosome 4. In contrast, they were asyntenic with 21 other enzymes including mouse dipeptidase-1 (DIP-1, human PEP-C; EC 3.4.11.(*), McKusick no. 17000) assigned to human chromosome arm 1q and mouse chromosome 1. Karyologic analysis confirmed this assignment. These data demonstrate that a large autosomal region (21 map units in the mouse and 51 map units in the human male) has been conserved in the evolution of mouse chromosome 4 and the short arm of human chromosome 1. Identification of such conserved regions will contribute to our understanding of the evolution of the mammalian genome and could suggest gene location by homology mapping.

    View details for Web of Science ID A1978FB94300072

    View details for PubMedID 209463

  • ASSIGNMENT OF THE GENES-CODING FOR PYROPHOSPHATASE AND HEXOKINASE-1 TO MOUSE CHROMOSOME-10 - IMPLICATIONS FOR COMPARATIVE GENE-MAPPING IN MAN AND MOUSE CYTOGENETICS AND CELL GENETICS Lalley, P. A., FRANCKE, U., Minna, J. D. 1978; 22 (1-6): 570-572

    View details for Web of Science ID A1978GU98300112

    View details for PubMedID 752542

  • REGIONAL MAPPING OF BETA-GLUCURONIDASE (BETAGUS) ON HUMAN CHROMOSOME-7 CYTOGENETICS AND CELL GENETICS George, D. L., FRANCKE, U. 1978; 22 (1-6): 437-440

    View details for Web of Science ID A1978GU98300078

    View details for PubMedID 752520

  • QUANTITATIVE-ANALYSIS OF HIGH-RESOLUTION TRYPSIN-GIEMSA BANDS ON HUMAN PROMETAPHASE CHROMOSOMES HUMAN GENETICS FRANCKE, U., OLIVER, N. 1978; 45 (2): 137-165

    Abstract

    We have constructed ideograms of human prometaphase chromosomes from synchronized and from standard 72-h lymphocyte cultures. G banding was achieved by a trypsin-Giemsa (or Wright's stain) method. In addition to light (white) and dark (black) bands, we have distinguished three different shades of grey. This distinction is essential for proper identification of the increasing number of bands displayed by high-resolution chromosomes. The relative amount of chromatin in each category of staining intensity has been calculated and expressed as 'light value.' The ideograms represent the maximal number of bands discernible with some consistency on prometaphase chromosomes, i.e., 721 euchromatic and 62 'variable' heterochromatic or heteromorphic bands. The ideograms are based on measurements. On selected printed copies of each chromosome derived from different cells and different individuals, the relative width of each band was measured in relation to the length of the respective chromosome arm. The measurements per chromosome were averaged and used for construction of the ideograms. The distance of each border between bands or sub-bands from the centromere has been calculated on a relative scale, with positions 0 at the centromere and 1.0 at the p terminus of q terminus. The numbering system for bands and sub-bands follows the Paris Conference (1971) recommendations.

    View details for Web of Science ID A1978GC07400006

    View details for PubMedID 738718

  • ASSIGNMENT OF A MUS-MUSCULUS GENE FOR TRIOSEPHOSPHATE ISOMERASE TO CHROMOSOME-6 AND FOR GLYOXALASE-I TO CHROMOSOME-17 USING SOMATIC-CELL HYBRIDS SOMATIC CELL GENETICS Minna, J. D., Bruns, G. A., KRINSKY, A. H., Lalley, P. A., FRANCKE, U., GERALD, P. S. 1978; 4 (2): 241-252

    Abstract

    Chinese hamster X mouse hybrid cells segregating mouse chromosomes have been used to assign a gene for triosephosphate isomerase (TPI-1, EC 5.3.1.1, McKusick No. 19045) to mouse chromosome 6, and a gene for Glyoxalase-I (GLO-1, EC 4.4.1.5, McKusick No 13875) to mouse chromosome 17. The genes for TPI-1 and lactate dehydrogenase B are syntenic in man and probably so in the dog. It is therefore likely that they are syntenic also in the mouse. It is of interest then that there is a mouse gene, Ldr-1, on chromosome 6 that regulates the level of LDH B subunits in mouse erythrocytes. The locus for GLO-1 is closely linked to the major histocompatibility complex in man. Since the major histocompatibility complex in the mouse is present on chromosome 17, this locus and the Glo-1 locus are syntenic in the mouse as well. This finding adds to the number of autosomal gene pairs which are syntenic in both mouse and man and reinforces the belief that there is considerable conservation. of linkage groups during evolution.

    View details for Web of Science ID A1978EV77600010

    View details for PubMedID 566476

  • REGIONAL ASSIGNMENT OF GENES FOR PHOSPHOGLUCOMUTASE-2 AND PEPTIDASE S TO 4PTER-]4Q21 IN MAN CYTOGENETICS AND CELL GENETICS FRANCKE, U., Brown, S. 1978; 22 (1-6): 401-405

    View details for Web of Science ID A1978GU98300069

    View details for PubMedID 752511

  • COMPARATIVE GENE-MAPPING IN MAN AND MOUSE - ASSIGNMENT OF THE GENES FOR LACTATE DEHYDROGENASE-A, PEPTIDASE-D, AND ISOCITRATE DEHYDROGENASE-2 TO MOUSE CHROMOSOME-7 CYTOGENETICS AND CELL GENETICS Lalley, P. A., FRANCKE, U., Minna, J. D. 1978; 22 (1-6): 577-580

    View details for Web of Science ID A1978GU98300114

    View details for PubMedID 752544

  • MOUSE CHROMOSOME-5 CODES FOR ECOTROPIC MURINE LEUKEMIA-VIRUS CELL-SURFACE RECEPTOR NATURE OIE, H. K., Gazdar, A. F., Lalley, P. A., RUSSELL, E. K., Minna, J. D., DELARCO, J., TODARO, G. J., FRANCKE, U. 1978; 274 (5666): 60-62

    View details for Web of Science ID A1978FE80700049

    View details for PubMedID 566388

  • HAGEMAN (FACTOR-XII) LOCUS ON 7Q - REPORT OF A 2ND CASE WITH DEL(7)Q35 AND NORMAL FACTOR-XII LEVEL HUMAN GENETICS FRANCKE, U. 1978; 45 (3): 363-367

    View details for Web of Science ID A1978GE25500014

    View details for PubMedID 738736

  • IDEOGRAMS OF HIGH-RESOLUTION TRYPSIN-GIEMSA BANDED HUMAN-CHROMOSOMES CYTOGENETICS AND CELL GENETICS OLIVER, N., FRANCKE, U. 1978; 22 (1-6): 668-671

    View details for Web of Science ID A1978GU98300136

    View details for PubMedID 752563

  • GENES-CODING FOR PYRUVATE-KINASE (M2) AND MANNOSEPHOSPHATE ISOMERASE ARE LINKED IN MAN AND MOUSE CYTOGENETICS AND CELL GENETICS Lalley, P. A., FRANCKE, U., Minna, J. D. 1978; 22 (1-6): 581-584

    View details for Web of Science ID A1978GU98300115

    View details for PubMedID 752545

  • Answer to criticism of Morton and Lalouel. American journal of human genetics FRANCKE, U., Felsenstein, J., Gartler, S. M., Nyhan, W. L., Seegmiller, J. E. 1977; 29 (3): 307-311

    View details for PubMedID 17948539

  • INVERTED TANDEM (MIRROR) DUPLICATIONS IN HUMAN-CHROMOSOMES - INV DUP 8P, 4Q, 22Q AMERICAN JOURNAL OF MEDICAL GENETICS Taylor, K. M., FRANCKE, U., Brown, M. G., George, D. L., Kaufhold, M. 1977; 1 (1): 3-19

    Abstract

    We have studied 4 patients with inverted tandem duplications of parts of chromosomes, a hitherto rarely identified form of a structural rearrangement involving a single chromosome in man. In patients 1 and 2, the duplication involved parts of the short arm of chromosome 8 (regions 8p12 leads to 8p23 and 8p21 leads to 8p23, respectively). Both patients manifested certain characteristics of the mosaic trisomy 8 syndrome. Elevated levels of glutathione reductase (GSR) in their erythrocytes supported the interpretation of a partial duplication of chromosome 8 and indicated a regional localization for the GSR gene locus. In Partient 3, the distal half of the long arm of chromosome 4 was duplicated (region 4q23 leads to 4q35). Clinical evidence supported this interpretation, as Patient 3 resembled phenotypically the 13 reported cases with duplication of the distal 4q. The cytogenetic findings in Patient 4 suggested a possibly inverted duplication of 22q. The clinical correlation was less convincing due to the lack of a well-defined phenotype for trisomy 22. These chromosome aberrations had occurred de novo in all 4 cases. Although they involved different chromosomal regions, they might well have arisen by the same mechanism. Possible modes of origin that are discussed in detail include unequal exchange between homologous chromosomes, between chromatids of 1 chromosome or between strands of 1 DNA duplex.

    View details for Web of Science ID A1977DY86000001

    View details for PubMedID 610424

  • IDENTIFICATION OF MOUSE CHROMOSOMES REQUIRED FOR MURINE LEUKEMIA-VIRUS REPLICATION CELL Gazdar, A. F., OIE, H., LALLEY, P., MOSS, W. W., Minna, J. D., FRANCKE, U. 1977; 11 (4): 949-956

    View details for Web of Science ID A1977DS42300021

    View details for PubMedID 196770

  • FAMILIAL TRISOMY 20P 5 CASES AND 2 CARRIERS IN 3 GENERATIONS - REVIEW ANNALES DE GENETIQUE CENTERWALL, W., FRANCKE, U. 1977; 20 (2): 77-83

    Abstract

    A clinically normal mother of three retarded children has been determined by G-banding to have a balanced translocation 46,XX,t(13;20) (q34;p11.2). The children each have an unbalanced form of the translocation with partial trisomy for 20p. Extensive gene marker studies have been unable to affix any specific gene locus onto the short arm of chromosome 20. The balanced translocation was inherited from the maternal grandfather. Two phenotypically abnormal deceased members of the family are believed to have had the unbalanced trisomy 20p condition. An increases number of spontaneous abortions were possibly due to lethal unbalanced 20p deletions. The moderate to mild mental retardation and somewhate unusual features (round face, prominent cheeks and nose, short mandible) in the three siblings and two other affected relatives suggest that trisomy of the short arm of chromosome 20 may cause a distinguishable clinical syndrome. Vertebral abnormalities and abnormal dermatoglyphics are part of the picture. Clinical and cytogenetic findings of all reported cases are compared.

    View details for Web of Science ID A1977DR72800001

    View details for PubMedID 302689

  • ASSIGNMENT OF MAJOR HISTOCOMPATIBILITY COMPLEX TO A REGION OF SHORT ARM OF HUMAN CHROMOSOME-6 PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA FRANCKE, U., Pellegrino, M. A. 1977; 74 (3): 1147-1151

    Abstract

    Interspecific cell hybrids containing defined parts of human chromosome 6 were used for regional mapping of gene loci previously assigned to chromosome 6: the human leukocyte antigens (HLA) region, phosphoglucomutase-3 (PGM3; alpha-D-glucose-1,6-bisphosphate:alpha-D-glucose-1-phosphate phosphotransferase, EC 2.7.5.1) and malic enzyme-1 [malic dehydrogenase(decarboxylating) (NADP+), L-malate:NADP+ oxidoreductase (oxaloacetate-decarboxylating), EC 1.1.1.40]. Human fibroblasts containing a balanced reciprocal translocation between the short arms of chromosome 1 and chromosome 6--designated t(1;6) (p3200;p2100)--were fused with an established line of Chinese hamster cells. Hybrid clones with segregating human chromosomes were studied for the presence of the translocation chromosomes 1T and 6T and their normal homologues 1 and 6. Six clones that had retained 1T, three clones with 6T, and three clones with 6 and 6T as controls, were analyzed by a microabsorption test for expression of the allelic antigens HLA-A2 and HLA-A3, both of which were present on the human parental cells. HLA-A2 segregated with the 1T translocation chromosome and HLA-A3 with the normal chromosome 6. Hybrid clones with 6T did not possess an HLA-A gene. In contrast, human PGM3 and malic enzyme-1 expression segregated with the 6T translocation chromosome. These results indicate the location of the major histocompatibility complex in region 6p2100 leads to 6pter of the short arm of chromosome 6. The genes for PGM2 and malic enzyme-1 map in region 6p2100 leads to 6qter. The HLA:PGM3 genetic map distance is 15 centimorgans in males, as established by family studies. This allows rather precise mapping of both loci because HLA is distal and PGM3 proximal to the translocation breakpoint in band 6p2100. The findings are consistent with earlier conclusions that HLA is proximal to 6p22. Quantitative correlation between the density of HLA antigens on the hybrid cell surface and the number of copies of the respective HLA gene-bearing chromosome suggests a gene dose effect for cell surface molecules, as it exists for intracellular gene products.

    View details for Web of Science ID A1977CZ32700075

    View details for PubMedID 265561

  • Experience with detection of heterozygous carriers and prenatal diagnosis of Lesch-Nyhan disease. Advances in experimental medicine and biology BAKAY, B., FRANCKE, U., Nyhan, W. L., Seegmiller, J. E. 1977; 76A: 351-358

    View details for PubMedID 855715

  • REGIONAL MAPPING OF HUMAN GENES FOR HEXOSAMINIDASE-B AND DIPHTHERIA-TOXIN SENSITIVITY ON CHROMOSOME-5 USING MOUSE X HUMAN HYBRID-CELLS SOMATIC CELL GENETICS George, D. L., FRANCKE, U. 1977; 3 (6): 629-638

    Abstract

    Mouse 3T3 (TK-) cells were fused to human leukocytes containing a balanced translocation [ins(3;5) (q27;q13q15)] in which part of the long arm of a chromosome 5 has been inserted into the long arm of a chromosome 3. Two independent, primary hybrid clones (XVI-10C;XVI-18A) retained the deleted chromosome 5 [del(5) (q13q15)] translocation product and were informative for regional mapping on chromosome 5 of genes involved in expression of hexosaminidase B (HEXB) and diphtheria toxin sensitivity (DTS). Both XVI-10C and XVI-18A clones were sensitive to diphtheria toxin. Toxin-resistant derivatives of these clones (XVI-10C DTR; XVI-18A DTR) were analyzed for chromosome content and expression of Hex B activity, as were XVI-10C and XVI-18A cells which had not been exposed to diphtheria toxin. The results of this study provide evidence for localization of DTS to region 5q15 leads to 5 qter on the long arm of chromosome 5, and localization of HEXB to region 5pter leads to 5q13.

    View details for Web of Science ID A1977ED72500006

    View details for PubMedID 601683

  • INTRACHROMOSOMAL GENE MAPPING IN MAN - GENE FOR TRYPTOPHANYL-TRANSFER-RNA SYNTHETASE MAPS IN REGION Q21-]QTER OF CHROMOSOME-14 SOMATIC CELL GENETICS FRANCKE, U., Denney, R. M., Ruddle, F. H. 1977; 3 (4): 381-389

    Abstract

    A gene for tryptophanyl-tRNA synthetase (EC 6.1.1.2), the enzyme which attaches tryptophan to its tRNA, has previously been assigned to human chromosome 14 by analysis of man-mouse somatic cell hybrids. We report here a method for the electrophoretic separation of Chinese hamster and human tryptophanyl-tRNA synthetases and its application to a series of independently derived Chinese hamster-human hybrids in which part of the human chromosome 14 has been translocated to the human X chromosome. When this derivative der (X),t(X;14) (Xqter leads to Xp22::14q21 leads to 14qter) chromosome carrying the human gene for hypoxanthine-guanine phosphoribosyltransferase was selected for and against in cell hybrid lines by the appropriate selective conditions, the human tryptophanyl-tRNA synthetase activity was found to segregate concordantly. These results provide additional confirmation for the assignment of the tryptophanyl-tRNA synthetase gene to human chromosome 14 and define its intrachromosomal location in the region 14q21 leads to 14qter. Our findings indicate that the genes for tryptophanyl-tRNA synthetase and for ribosomal RNA are not closely linked on chromosome 14.

    View details for Web of Science ID A1977DQ96600003

    View details for PubMedID 564085

  • CONGENITAL ANOMALIES INCLUDING VATER ASSOCIATION IN A PATIENT WITH A DEL(6)Q DELETION JOURNAL OF PEDIATRICS MCNEAL, R. M., FRANCKE, U., SKOGLUND, R. R. 1977; 91 (6): 957-960

    View details for Web of Science ID A1977EC66200026

    View details for PubMedID 925830

  • GENE DOSE EFFECT - INTRABAND MAPPING OF LDH-A LOCUS USING CELLS FROM 4 INDIVIDUALS WITH DIFFERENT INTERSTITIAL DELETIONS OF 11P CYTOGENETICS AND CELL GENETICS FRANCKE, U., George, D. L., Brown, M. G., Riccardi, V. M. 1977; 19 (4): 197-207

    Abstract

    The quantitative expression of LDH A was studied in hemolysates from four patients with different but overlapping interstitial deletions of the short arm of chromosome 11. Deficiency of LDH A was demonstrated in one patient, and the LDH A locus has been assigned to that segment of 11p for which this patient alone was deficient, i.e., to band 11p12 (region 11p1203 leads to 11p1208).

    View details for Web of Science ID A1977EH94000002

    View details for PubMedID 598250

  • PARTIAL 4Q DUPLICATION DUE TO INHERITED DER(20), T(4-20)(Q25-Q13)MAT ANNALES DE GENETIQUE Sparkes, R. S., FRANCKE, U., Muller, H., Toomey, K. 1977; 20 (1): 31-35

    Abstract

    A boy with mental and growth retardation associated with congenital anomalies has a partial duplication of the distal 4q chromosome region as a result of inheritance of a t(4:20) from his mother. Comparison with twelve other patients from the literature indicates that similar clinical features may be associated with this chromosome change suggesting a partial 4q duplication syndrome.

    View details for Web of Science ID A1977DD72000005

    View details for PubMedID 302670

  • GENE MAPPING IN MUS-MUSCULUS BY INTERSPECIFIC CELL HYBRIDIZATION - ASSIGNMENT OF GENES FOR TRIPEPTIDASE-1 TO CHROMOSOME-10, DIPEPTIDASE-2 TO CHROMOSOME-18, ACID PHOSPHATASE-1 TO CHROMOSOME-12, AND ADENYLATE KINASE-1 TO CHROMOSOME-2 CYTOGENETICS AND CELL GENETICS FRANCKE, U., Lalley, P. A., Moss, W., Ivy, J., Minna, J. D. 1977; 19 (2-3): 57-84

    Abstract

    Chinese hamster X mouse somatic cell hybrids segregating mouse chromosomes were examined for their mouse chromosome content using trypsin-Giemsa (GTG) banding and Hoechst 33258 staining techniques. Simultaneously, they were scored for the presence of 24 mouse enzymes. The results confirm the assignments of 11 genes previously mapped by sexual genetics: Dip-1 and Id-1 to chromosome 1; Pgm-2 and Pgd to 4; Pgm-1 to 5; Gpi-1 to 7; Gr-1 to 8; Mpi-1 and Mod-1 to 9; Np-1 and Es-10 to 14. They also confirm chromosomally the assignments of 3 genes that were made by other somatic cell genetic studies: Aprt to 8; Hprt and alpha-gal to the X chromosome. But most importantly, four enzyme loci are assigned to four chromosomes that until now were not known to carry a biochemical marker which is expressed in cultured cells: Trip-1 to 10; Dip-2 to 18; Acp-1 to 12; and Ak-1 to 2. Cytogenetic examination of clones showing discordant segregation of HPRT and A-GAL, suggested the assignment of alpha-gal to region XE leads to XF of the mouse X chromosome. The cytologic studies provide a comparison between data from sexual genetics and somatic cell hybrids and validate hybrid cell techniques. They provide evidence of the reliability of scoring chromosomes by GTG and Hoechst staining and stress the importance of identifying clones with multiple chromosome rearrangements. Striking examples of norandom segregation of mouse chromosomes were observed in these hybrids with preferential retention of 15 and segregation of 11 and the Y chromosome.

    View details for Web of Science ID A1977DY39800001

    View details for PubMedID 198184

  • Duplication 11 (q21 to 23 leads to qter) syndrome. Birth defects original article series FRANCKE, U., Weber, F., Sparkes, R. S., MATTSON, P. D., Mann, J. 1977; 13 (3B): 167-186

    View details for PubMedID 890090

  • The pallister mosaic syndrome. Birth defects original article series PALLISTER, P. D., Meisner, L. F., Elejalde, B. R., FRANCKE, U., Herrmann, J., Spranger, J., Tiddy, W., Inhorn, S. L., Opitz, J. M. 1977; 13 (3B): 103-110

    View details for PubMedID 890087

  • PROXIMAL 4P-DELETION - PHENOTYPE DIFFERS FROM CLASSICAL 4P-SYNDROME JOURNAL OF PEDIATRICS FRANCKE, U., Arias, D. E., Nyhan, W. L. 1977; 90 (2): 250-252

    View details for Web of Science ID A1977CU27100018

    View details for PubMedID 830918

  • Cytogenetics and somatic cell genetics: the impact of chromosome banding. Birth defects original article series FRANCKE, U. 1977; 13 (6): 79-103

    View details for PubMedID 72574

  • PARTIAL TRISOMY-20P DERIVED FROM A T(18-20) TRANSLOCATION HUMAN GENETICS Taylor, K. M., WOLFINGER, H. L., Brown, M. G., Chadwick, D. L., FRANCKE, U. 1976; 34 (2): 155-162

    Abstract

    Two sibs show a strikingly concordant syndrome of congenital anomalies and G-banding reveals that each has partial trisomy 20p resulting from a t(18;20) translocation. They resemble other cases of partial trisomy 20p in some respects but also differ in some ways. Their normal sib, mother, and half-aunt are balanced heterozygotes for the t(18;20) translocation. The segregation of the balanced translocation in this family is associated with an extremely poor reproductive record. The segregation pattern closely parallels that of a t(13;20) translocation in a family described by Carrel et al. (1971) and Francke (1972). The similarity of segregation patterns is predictable on the basis of probable pachytene configurations, but the dissimilarity of phenotypes between families is not readily explained.

    View details for Web of Science ID A1976CK04000006

    View details for PubMedID 1002139

  • GENE DOSE EFFECT - REGIONAL MAPPING OF HUMAN NUCLEOSIDE PHOSPHORYLASE ON CHROMOSOME 14 SCIENCE George, D. L., FRANCKE, U. 1976; 194 (4267): 851-852

    Abstract

    Quantitative analyses of erythrocyte nucleoside phosphorylase in four unrelated cases of partial trisomy 14 indicate that the structural gene for this enzyme maps in the chromosome region 14q11 leads to 14q21.

    View details for Web of Science ID A1976CK55500024

    View details for PubMedID 824731

  • HUMAN GENE FOR BETA-GLUCURONIDASE IS ON CHROMOSOME-7 AMERICAN JOURNAL OF HUMAN GENETICS FRANCKE, U. 1976; 28 (4): 357-362

    Abstract

    Inconsistent assignments of the human gene for beta glucuronidase (GUS) to chromosomes 7 and 9 have previously been reported. In this study, we have correlated the expression of human GUS in 22 primary Chinese hamster/human hybrid lines with quantitative cytogenetic analysis. Eight hybrids were positive for human GUS as manifested by a five-band pattern on electrophoresis. All of them contained a human chromosome 7 in 34% or more of cells, and seven of them had not retained chromosome 9. One hybrid with only 6% of metaphases possessing a chromosome 7 had no detectable human GUS activity. Human GUS expression was absent in 10 hybrid clones containing chromosome 9 but not 7 and in control fibroblasts from a patient with GUS deficiency. These results support the assignment of presumably the structural gene for beta glucuronidase to chromosome 7.

    View details for Web of Science ID A1976BX34500005

    View details for PubMedID 941903

  • INTRACHROMOSOMAL GENE MAPPING IN MAN - ASSIGNMENT OF NUCLEOSIDE PHOSPHORYLASE TO REGION 14CEN-]14Q21 BY INTERSPECIFIC HYBRIDIZATION OF CELLS WITH A T(X-14) (P22-Q21) TRANSLOCATION SOMATIC CELL GENETICS FRANCKE, U., BUSBY, N., Shaw, D., Hansen, S., Brown, M. G. 1976; 2 (1): 27-40

    Abstract

    The structural gene for purine-nucleoside phosphorylase (NP) has been assigned to a subregion of chromosome 14 by somatic cell hybridization of male and female cells containing the balanced translocation t(X;14) (p22;q21). Peripheral lymphocytes were fused to a pseudodiploid HPRT-deficient established Chinese hamster cell line. 23 primary hybrid clones (10 derived from male and 13 from female cells) were isolated and maintained in HAT selective medium. Parallel subcultures from generations 16, 24, and 40 after clonal isolation were fully karyotyped and analyzed electrophorectically for expression of the human types of NP, HPRT, G6PD, and PGK. The human NP phenotype segregated discordantly with each human chromosome except chromosome 14 and the der(14),t(X;14) translocation chromosome. In all, 8 hybrids which had retained the der(X), t(X;14) translocation chromosome under HAT selective pressure and expressed human HPRT had lost the human NP phenotype. These results indicate localization of the NP gene in region 14pter leads to 14q21.

    View details for Web of Science ID A1976BL99600004

    View details for PubMedID 829289

  • COMPARATIVE MAPPING USING SOMATIC-CELL HYBRIDS IN VITRO-JOURNAL OF THE TISSUE CULTURE ASSOCIATION Minna, J. D., Lalley, P. A., FRANCKE, U. 1976; 12 (11): 726-733

    Abstract

    Comparative mapping, or ascertaining the gene linkage relationships between different species, is rapidly developing. This is possible because new techniques in chromosome identification and somatic cell hybridization, such as the generation of hybrids preferentially segregating chromosomes of any desired species including rodents, and the development of gene transfer techniques have yielded new information about the human and rodent gene maps. In addition, the discovery and characterization of mouse subspecies has generated new mouse sexual genetic linkage data. The following picture is emerging. Several X-linked genes in man are X-linked in all mammalian species tested. The linkage relationships of several tightly linked genes, less than 1 map unit apart, are also conserved in all mammalian species tested. Ape autosomal genes are assigned to ape chromosomes homologous to their human counterparts indicating extensive conservation in the 12 million years (MYR) of evolution from apes to man. Similarly, mouse and rat, 10 MYR apart in evolution, have several large autosomal synteny groups conserved. In comparing the mouse and human gene maps we find that human genes assigned to different arms of the same human chromosome are unlinked in the mouse; mouse genes large map distances (20 to 45 cM) apart are very likely to be unlinked in the human. However, several autosomal synteny groups 10 to 20 cM apart, including the Pgd, Eno-1, Pgm-1 group on human chromosome arm 1p, are conserved in mice and man. This suggests that homology mapping, the superimposition of one species gene map on the homologous conserved portion of another species genome may be possible, and that ancestral autosomal synteny groups should be detectable.

    View details for Web of Science ID A1976CY37100003

    View details for PubMedID 828614

  • ASSIGNMENT OF NUCLEOSIDE-PHOSPHORYLASE-(NP) GENE LOCUS TO REGION 14PTER-]14Q21 CYTOGENETICS AND CELL GENETICS FRANCKE, U., BUSBY, N., Shaw, D., Hansen, S., Brown, M. G. 1976; 16 (1-5): 135-137

    View details for Web of Science ID A1976CK04200023

    View details for PubMedID 824099

  • OCCURRENCE OF NEW MUTANTS IN X-LINKED RECESSIVE LESCH-NYHAN DISEASE AMERICAN JOURNAL OF HUMAN GENETICS FRANCKE, U., Felsenstein, J., Gartler, S. M., Migeon, B. R., Dancis, J., Seegmiller, J. E., BAKAY, F., Nyhan, W. L. 1976; 28 (2): 123-137

    Abstract

    In a population at equilibrium for a sex-linked lethal, one-third of the genes for that lethal must arise anew each generation. Therefore, one-third of all cases of Lesch-Nyhan disease, a severe X-linked recessive lethal disorder, should be new mutants. To test this hypothesis, we have collected 47 families, 20 with a single proband and 27 with multiple affected males in which the patients' mothers and other female relatives had been studied for heterozygosity. Available carrier detection tests identify heterozygous for HPRT deficiency in hair roots and skin fibroblasts. Only four mothers were found not to be carriers. This result deviates significantly from expected (P less than .001). Statistical tests for ascertainment effects indicated absence of bias for multiple proband families but strong bias in favor of families with many heterozygous females. When the analysis was limited to single proband families, the deviation from expected was still significant (P less than .01). The incidence of new mutants among the heterozygous mothers, as determined by the ratio of +/+ to +/- maternal grandmothers, should be one-half (see Appendix). Of all 20 maternal grandmothers studied, five were +/+ and 15 were +/- (P less than .05). Considering only the single proband families, the ratio of 5 +/+ to 8 +/- was not significantly different from expected. In four of the five cases in which the heterozygous mother of an affected individual was a new mutation, the age of her parents was considerably higher than the mean parental age in the population. This raises the possibility of a paternal age effect on X-linked mutations. There appears to be a true deficiency of new mutatnts among males but not among females. Data on additional Lesch-Nyhan families are needed before conclusions regarding a possible higher mutation rate in males can be drawn.

    View details for Web of Science ID A1976BJ47600002

    View details for PubMedID 1266847

  • The gene for the species antigen on human chromosome 11 is on the short arm. Birth defects original article series Buck, D. W., Bodmer, W. F., BOBROW, M., FRANCKE, U. 1976; 12 (7): 97-98

    View details for PubMedID 1024660

  • REGIONAL ASSIGNMENTS OF GENES FOR FUMARATE HYDRATASE AND GUANYLATE KINASE ON CHROMOSOME-1 AND FOR LYSOSOMAL ACID-PHOSPHATASE AND ESTERASE A4 ON CHROMOSOME-11 CYTOGENETICS AND CELL GENETICS BUSBY, N., COURVAL, J., FRANCKE, U. 1976; 16 (1-5): 105-107

    View details for Web of Science ID A1976CK04200014

    View details for PubMedID 185008

  • Regional assignments of the genes for fumarate hydratase and guanylate kinase on chromosome 1 and for lysosomal acid phosphatase and esterase A4 on chromosome 11. Birth defects original article series BUSBY, N., COURVAL, J., FRANCKE, U. 1976; 12 (7): 105-107

    View details for PubMedID 1024600

  • RETINOBLASTOMA AND CHROMOSOME-13 CYTOGENETICS AND CELL GENETICS FRANCKE, U. 1976; 16 (1-5): 131-134

    View details for Web of Science ID A1976CK04200022

    View details for PubMedID 975871

  • GENE DOSE EFFECT - REGIONAL MAPPING OF HUMAN GLUTATHIONE REDUCTASE ON CHROMOSOME-8 CYTOGENETICS AND CELL GENETICS George, D. L., FRANCKE, U. 1976; 17 (5): 282-286

    View details for Web of Science ID A1976CW21600004

    View details for PubMedID 1017318

  • 2P PARTIAL TRISOMY SYNDROME - DUPLICATION OF REGION 2P23-]2PTER IN 2 MEMBERS OF A T(2-7) TRANSLOCATION KINDRED AMERICAN JOURNAL OF DISEASES OF CHILDREN FRANCKE, U., Jones, K. L. 1976; 130 (11): 1244-1249

    Abstract

    Strikingly similar abnormalities were present in two severely retarded children, an 8-year-old boy and a 12-year-old girl, who were first cousins once removed. Dysmorphic features included high bulging forehead with frontal upsweep of hair, flat, wide glabella and nasal bridge, maxillary hypoplasia, ptosis, dacryostenosis, dolichostenomelia, hyperextensible fingers with subluxation in proximal interphalangeal joints, hypoplastic external genitalia, and overconstriction of the shafts of all long bones. Both patients had unbalanced karyotypes, with duplication of the distal third of the short arm of chromosome 2, resulting from the same balanced reciprocal translocation prestne in both mothers: der (2), t(2;7)(p23;q36)mat. Seven additional translocation carriers were identified in four generations of this kindred.

    View details for Web of Science ID A1976CL18300014

    View details for PubMedID 984008

  • GENE FOR SPECIES ANTIGEN ON HUMAN CHROMOSOME-11 IS ON SHORT ARM CYTOGENETICS AND CELL GENETICS Buck, D. W., Bodmer, W. F., BOBROW, M., FRANCKE, U. 1976; 16 (1-5): 97-98

    View details for Web of Science ID A1976CK04200011

    View details for PubMedID 61840

  • NEW SYNDROME OF MENTAL DEFICIENCY WITH CRANIOFACIAL, LIMB, AND ANAL ABNORMALITIES JOURNAL OF PEDIATRICS Keller, M. A., Jones, K. L., Nyhan, W. L., FRANCKE, U., Dixson, B. 1976; 88 (4): 589-591

    View details for Web of Science ID A1976BL22500012

    View details for PubMedID 1255317

  • SPORADIC BILATERAL RETINOBLASTOMA AND 13Q-CHROMOSOMAL DELETION MEDICAL AND PEDIATRIC ONCOLOGY FRANCKE, U., Kung, F. 1976; 2 (4): 379-385

    Abstract

    Unilateral retinoblastoma (Rb) is usually a sporadic occurrence while bilateral (multifocal) cases are often familial. Sporadic bilateral Rb associated with a long-arm deletion of a D-group chromosome has been reported in 8 children. We have studied a 6-year-old female with bilateral sporadic retinoblastoma, treated during infancy by enucleation and radiotherapy. Chromosome banding studies on peripheral lymphocytes revealed an interstitial deletion from the long arm of a chromosome 13: del(13) (q12q14). Three additional patients reported in the literature had interstitial 13q- deletions, involving slightly different though overlapping regions. The only chromosomal region consistently missing in all of these 4 cases appears to be part of the lightly staining band 13q14. We, therefore, propose this site as the precise location of a gene (or genes) involved in retinal development. Our patient lacked features of the classic 13q- or 13-ring syndrome, which involves deletion of a more distal portion of the 13 long arm. When compared to reported patients with Rb and 13q-, it became apparent that there may be a separate recognizable syndrome consisting of moderate growth and developmental delay, characteristic facies and external ears, and bilateral sporadic Rb, which is associated with an interstitial 13q- deletion.

    View details for Web of Science ID A1976CR42300003

    View details for PubMedID 1004381

  • Retinoblastoma and chromosome 13. Birth defects original article series FRANCKE, U. 1976; 12 (7): 131-134

    View details for PubMedID 1024606

  • DEL(10)P AUTOSOMAL DELETION SYNDROME - CLINICAL, CYTOGENETIC AND GENE MARKER STUDIES HUMANGENETIK FRANCKE, U., KERNAHAN, C., Bradshaw, C. 1975; 26 (4): 343-351

    Abstract

    A 46,XX,del(10)p13 karyotype (Paris Conference, 1971) was identified in a 5-year-old Negro girl with mental and growth retardation, brachy- and trigonocephaly, downward slanting palpebral fissures, hypotelorism, epicanthal folds, ptosis, strabismus, dysplastic nose, high-arched palate, microdontia, small low-set posteriorly rotated ears, asymmetrical thorax, wide-spaced nipples, and minor abnormalities of hands and feet. Both parents and a brother had normal karyotypes. Expression of more than 50 polymorphic gene loci determining blood groups, serum proteins and red cell enzymes was studied. The results did not permit localization of a gene locus on the deleted segment of chromosome 10. The proposita was heterozygous for the Rh and MN blood groups and for the red cell enzymes adenosine deaminase, glutamate pyruvate transaminase and esterase D. These gene loci are thereby excluded from region 10p13 yields 10pter.

    View details for Web of Science ID A1975V798700008

    View details for PubMedID 1150232

  • ASSIGNMENTS OF HUMAN GENES FOR LACTATE DEHYDROGENASE-A AND THYMIDINE KINASE TO SPECIFIC CHROMOSOMAL REGIONS CYTOGENETICS AND CELL GENETICS FRANCKE, U., BUSBY, N. 1975; 14 (3-6): 313-319

    View details for Web of Science ID A1975AP45700028

    View details for PubMedID 1192784

  • Trisomy 14q-. Birth defects original article series Fawcett, W. A., MCCORD, W. K., FRANCKE, U. 1975; 11 (5): 223-228

    Abstract

    Trisomy 14q- syndrome is relatively new and needs further delineation. In comparing our case with other reported cases (Table 1), some similarities are seen. Although the comparison of our case of partial trisomy 14 with the other cases reported in the literature may not be entirely justified (in that the reciprocal translocations are not always identical), it is hoped that by doing so, we can further delineate the common features and prognosis of such individuals.

    View details for PubMedID 1218218

  • 10p-: a new autosomal deletion syndrome? Birth defects original article series FRANCKE, U., Mahan, G. M., DIXSON, B. K., Jones, O. W. 1975; 11 (5): 207-212

    View details for PubMedID 1218215

  • REGIONAL LOCALIZATION OF HUMAN GENES FOR MALATE DEHYDROGENASE-1 AND ISOCITRATE DEHYDROGENASE-1 ON CHROMOSOME-2 BY INTERSPECIFIC HYBRIDIZATION USING HUMAN CELLS WITH BALANCED RECIPROCAL TRANSLOCATION T(1-2) (Q32-Q13) CYTOGENETICS AND CELL GENETICS FRANCKE, U. 1975; 14 (3-6): 308-312

    View details for Web of Science ID A1975AP45700027

    View details for PubMedID 1192808

  • HALF CHROMATID MUTATIONS - TRANSMISSION IN HUMANS AMERICAN JOURNAL OF HUMAN GENETICS Gartler, S. M., FRANCKE, U. 1975; 27 (2): 218-223

    Abstract

    Attention is drawn to the possibility of half chromatid and early somatic mutations and to several implications of these mosaic-yielding events. There is suggestive evidence that spontaneous mutations can result in mosaics. A world-wide cooperative study of Lesch-Nyhan families could determine the extent of half chromatid mutation transmission and early somatic mutation in humans.

    View details for Web of Science ID A1975V946700008

    View details for PubMedID 1124765

  • Assignments of the human genes for lactate dehydrogenase-A and thymidine kinase to specific chromosomal regions. Birth defects original article series FRANCKE, U., BUSBY, N. 1975; 11 (3): 143-149

    View details for PubMedID 1203473

  • PRENATAL DIAGNOSIS OF TRISOMY 9 HUMANGENETIK FRANCKE, U., Benirschke, K., Jones, O. W. 1975; 29 (3): 243-250

    Abstract

    A male karyotype with trisomy 9 (47,XY,+9) was identified in amniotic fluid cells from a 40-year-old pregnant woman. After termination of the pregnancy by saline abortion, the cytogenetic diagnosis was confirmed in a cell line grown from placental tissue. Microscopic and gross pathological findings in the fetus were compared to 1 case with trisomy 9 and 3 with trisomy 9 mosaicism reported in the literature. A distinct clinical syndrome appears to be associated with this rare autosomal trisomy.

    View details for Web of Science ID A1975AS13200009

    View details for PubMedID 1165103

  • Translocation, t(4qminus;13qplus), in three generations resulting in partial trisomy of the long arm of chromosome 4 in the fourth generation. Journal of medical genetics Schrott, H. G., Sakaguchi, S., FRANCKE, U., LUZZATTI, L., FIALKOW, P. J. 1974; 11 (2): 201-205

    View details for PubMedID 4135220

  • LINKAGE RELATIONSHIPS OF X-LINKED ENZYMES GLUCOSE-6-PHOSPHATE-DEHYDROGENASE AND HYPOXANTHINE GUANINE PHOSPHORIBOSYLTRANSFERASE - RECOMBINATION IN FEMALE OFFSPRING OF COMPOUND HETEROZYGOTES AMERICAN JOURNAL OF HUMAN GENETICS FRANCKE, U., BAKAY, B., Connor, J. D., Coldwell, J. G., Nyhan, W. L. 1974; 26 (4): 512-522

    View details for Web of Science ID A1974T505100009

    View details for PubMedID 4842775

  • TRANSLOCATION, T(4Q--13Q+), IN 3 GENERATIONS RESULTING IN PARTIAL TRISOMY OF LONG ARM OF CHROMOSOME 4 IN FOURTH GENERATION JOURNAL OF MEDICAL GENETICS Schrott, H. G., SAKAGUCH, S., FRANCKE, U., LUZZATTI, L., FIALKOW, P. J. 1974; 11 (2): 201-205
  • MODES OF GROWTH AND SPREAD OF A TRANSPLANTABLE, VIRUS-PRODUCING MURINE (MOLONEY) SARCOMA - KARYOTYPIC ANALYSES JOURNAL OF THE NATIONAL CANCER INSTITUTE Russell, S. W., FRANCKE, U., Buettner, L., Cochrane, C. G. 1974; 53 (3): 801-806

    View details for Web of Science ID A1974U218500029

    View details for PubMedID 4414698

  • PROPERTIES OF CYSTINOTIC FIBROBLAST-D98 CELL HYBRIDS STUDIED BY SOMATIC-CELL HYBRIDIZATION NATURE SCHNEIDE, J. A., FRANCKE, U., Hammond, D. S., PELLETT, O. L., BECKER, F. L. 1973; 244 (5414): 289-292

    View details for Web of Science ID A1973Q291300030

    View details for PubMedID 4583104

  • EVIDENCE AGAINST PERIPHERAL LOCATION OF Y-CHROMOSOME IN HUMAN METAPHASE CELLS CYTOGENETICS AND CELL GENETICS SPENCE, M. A., FRANCKE, U., FORSYTHE, A. B. 1973; 12 (1): 49-52

    View details for Web of Science ID A1973P723400006

    View details for PubMedID 4707247

  • BAND PATTERNS OF 12 D 98-AH-2 MARKER CHROMOSOMES AND THEIR USE FOR IDENTIFICATION OF INTRASPECIFIC CELL HYBRIDS CHROMOSOMA FRANCKE, U., Hammond, D. S., SCHNEIDE, J. A. 1973; 41 (1): 111-121

    View details for Web of Science ID A1973O789100009

    View details for PubMedID 4120821

  • DETECTION OF HETEROZYGOUS CARRIERS OF LESCH-NYHAN SYNDROME BY ELECTROPHORESIS OF HAIR ROOT LYSATES JOURNAL OF PEDIATRICS FRANCKE, U., BAKAY, B., Nyhan, W. L. 1973; 82 (3): 472-478

    View details for Web of Science ID A1973O919600015

    View details for PubMedID 4349227

  • SYSTEM OF NOMENCLATURE FOR BAND PATTERNS OF MOUSE CHROMOSOMES CHROMOSOMA NESBITT, M. N., FRANCKE, U. 1973; 41 (2): 145-158

    View details for Web of Science ID A1973P038000002

    View details for PubMedID 4120886

  • Karyotypic, virologic, and immunologic analyses of two continuous lymphocyte lines established from New Zealand black mice: possible relationship of chromosomal mosaicism to autoimmunity. Proceedings of the National Academy of Sciences of the United States of America Lerner, R. A., Jensen, F., Kennel, S. J., Dixon, F. J., Des Roches, G., FRANCKE, U. 1972; 69 (10): 2965-2969

    Abstract

    Two continuous-suspension lymphocyte lines were isolated from the spleen and fibrosarcoma of a New Zealand Black female mouse. A C-type virus with a density of 1.16 g x cm(-3), 70S RNA, and RNA-directed DNA polymerase activity was isolated. The virus was infectious for NRK, NZB, (NZB x NZW)F(1), and (NZW x NZW)F(1) embryos, and for BALB/c 3T3 cells, but not for NIH Swiss cells. All cells from established lymphocyte cultures, as well as some embryo cells from New Zealand Black mice, showed karyotypic abnormalities. The possibility of chromosomal mosaicism is suggested.

    View details for PubMedID 4342970

  • Cytological identification of the chromosomes involved in Searle's translocation and the location of the centromere in the X chromosome of the mouse. Genetics Eicher, E. M., NESBITT, M. N., FRANCKE, U. 1972; 71 (4): 643-648

    Abstract

    The autosome in Searle's X-autosome translocation has been shown to be chromosome 16. The breakpoint in chromosome 16 is slightly proximal to the middle and in the X is slightly distal to the middle.-Available evidence indicates that either Linkage Group XV or Linkage Group XIX is carried on chromosome 16.-The centromere of the X chromosome is at the spf end of the linkage group.

    View details for PubMedID 5055131

  • QUINACRINE MUSTARD FLUORESCENCE OF HUMAN CHROMOSOMES - CHARACTERIZATION OF UNUSUAL TRANSLOCATIONS AMERICAN JOURNAL OF HUMAN GENETICS FRANCKE, U. 1972; 24 (2): 189-?

    View details for Web of Science ID A1972M254100008

    View details for PubMedID 5016511

  • KARYOTYPIC, VIROLOGIC, AND IMMUNOLOGICAL ANALYSES OF 2 CONTINUOUS LYMPHOCYTE LINES ESTABLISHED FROM NEW-ZEALAND BLACK MICE - POSSIBLE RELATIONSHIP OF CHROMOSOMAL MOSAICISM TO AUTOIMMUNITY PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Lerner, R. A., Dixon, F. J., DESROCHE, G., Kennel, S. J., FRANCKE, U., Jensen, F. 1972; 69 (10): 2965-?
  • INHERITED T(13Q14Q) IN 2 RETARDED SISTERS AMERICAN JOURNAL OF HUMAN GENETICS Crandall, B. F., Sparkes, R. S., FRANCKE, U., Campbell, M. A. 1972; 24 (4): 416-?

    View details for Web of Science ID A1972M806300005

    View details for PubMedID 5031981

  • LINKAGE GROUP-II AND GROUP-12 OF MOUSE - CYTOLOGICAL LOCALIZATION BY FLUOROCHROME STAINING SCIENCE Nesbitt, M., FRANCKE, U. 1971; 174 (4004): 60-?

    Abstract

    The T(2;12) 163H translocation involves two chromosomes which carry the two groups of genes known as linkage groups II and XII. Staining with quinacrine mustard reveals that these chromosomes are numbers 10 and 19 of the fluorescent karyotype of the normal mouse. Linkage group II is assigned to chromosome 10, linkage group XII to chromosome 19.

    View details for Web of Science ID A1971K398100018

    View details for PubMedID 4107692

  • ANALYSIS OF T(3-QUESTIONABLE)6CA AND T(14-17)264CA TRANSLOCATIONS IN MOUSE BY QUINACRINE MUSTARD STAINING GENETICS Nesbitt, M., FRANCKE, U. 1971; 69 (4): 517-?

    View details for Web of Science ID A1971L494400009

    View details for PubMedID 4110444

  • CATTANACHS TRANSLOCATION - CYTOLOGICAL CHARACTERIZATION BY QUINACRINE MUSTARD STAINING PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA FRANCKE, U., Nesbitt, M. 1971; 68 (12): 2918-?

    Abstract

    Metaphase chromosomes of mice carrying Cattanach's translocation, which is the deletion of material from a medium-sized autosome and its insertion into an X chromosome, were stained with quinacrine mustard. Comparison of the fluorescence patterns of these chromosomes with those of the chromosomes of normal mice has allowed the identification of the autosome involved in the translocation and localization of the transposed material within the X chromosome. Since the material inserted into the X chromosome in Cattanach's translocation is known to carry part of linkage group I, we are now able to assign linkage group I to a specific chromosome pair of the normal fluorescent karyotype of the mouse.

    View details for Web of Science ID A1971L129000004

    View details for PubMedID 5289234

  • IDENTIFICATION OF MOUSE CHROMOSOMES BY QUINACRINE MUSTARD STAINING CYTOGENETICS FRANCKE, U., Nesbitt, M. 1971; 10 (5): 356-?

    View details for Web of Science ID A1971L116600005

    View details for PubMedID 4109887