Bio


Dr Anna Gloyn joined the faculty at Stanford University in February 2020 after sixteen years at the University of Oxford, UK. Dr Gloyn completed her DPhil at the University of Oxford under the supervision of the late Professor Robert Turner. Her post-doctoral training was carried out at the University of Exeter under the mentorship of Professors Andrew Hattersley & Sian Ellard and at the University of Pennsylvania in Philadelphia under the mentorship of Professor Franz Matschinsky. In 2004 she returned to Oxford with a Diabetes UK RD Lawrence Career Development Fellowship and established an independent research group focused on understanding beta-cell function through the investigation of genetic variants causally implicated in monogenic diabetes. In 2011 she was awarded a prestigious Wellcome Senior Fellowship in Basic Biomedical Science which she successfully renewed in 2016 and transferred to Stanford when she relocated.

Academic Appointments


Administrative Appointments


  • Associate Chair for Basic Science Research, Department of Pediatrics (2020 - Present)
  • Co-Director of the Pilot & Feasibility Program, Stanford Diabetes Research Center (2023 - Present)
  • Co-Director of Enrichment Program, Stanford Diabetes Research Center (2023 - Present)
  • Co-Director of Recruitment for Physician-Scientist Training Program, Pediatrics Residency Program (2022 - Present)
  • Co-Lead for Basic & Translational Science Scholarly Concentration, Pediatrics Residency Program (2020 - Present)
  • Co-Lead of Pancreas & Islet Affinity Group, Stanford Diabetes Research Centre (2020 - Present)
  • Member, Stanford Diabetes Research Centre (2020 - Present)

Honors & Awards


  • Outstanding Scientific Achievement Award, American Diabetes Association (June 2022)
  • Dorothy Hodgkin Named Lecture, Diabetes UK (March 2019)
  • G.B. Morgagni Prize Silver Medal, University of Padua Medical School, Padua, Italy (October 2014)
  • Minkowski Award, European Association for the Study of Diabetes (October 2014)
  • RD Lawrence Named Lecture, Diabetes UK (March 2009)
  • Rising Star Award, European Association for the Study of Diabetes (September 2005)

Boards, Advisory Committees, Professional Organizations


  • Associate Editor, Diabetes Care (2024 - Present)
  • Editorial Advisory Board, Cell Genomics (2021 - Present)
  • Chair of Working Group Precision Diagnostics for Monogenic Diabetes, American Diabetes Association Precision Medicine Consensus Working Group (2020 - Present)
  • Member of the Executive Committee, Atlas of Variant Effects Alliance (2020 - Present)
  • Member of the Flagship Disease (Diabetes) Working Group, International Common Disease Alliance (2020 - Present)
  • Member of the Working Group (Mechanisms), International Common Disease Alliance (2020 - Present)
  • Member of the Working Group (Medicines), International Common Disease Alliance (2019 - Present)
  • Panel Member, ClinGen Monogenic Diabetes Variant Curation Expert Panel (2019 - Present)
  • Associate Editor, Endocrine Reviews (2018 - 2021)
  • Editorial Advisory Board, Diabetologia (2018 - 2021)
  • Trustee, European Diabetology (2017 - 2021)
  • Expert Review Panel Member for Genetics Genomics & Population Health, Wellcome Trust (2016 - 2021)

Current Research and Scholarly Interests


The consistent focus of Anna’s research has been using naturally occurring mutations in humans as tools to identity critical regulatory pathways and insights into normal physiology. Her early post-doctoral research led to the identification a new genetic aetiology for permanent and transient neonatal diabetes due to KCNJ11 mutations and resulted in one of the first examples of precision medicine, where the determination of the molecular genetic aetiology lead to improved treatment options for patients. Whilst in Oxford, Anna's team discovered a novel genetic cause of constitutive insulin sensitivity in humans due to mutations in the PTEN gene highlighting the complex interplay between pathways involved in cell-growth and metabolism.

Her early independent work focused on the clinical and functional characterization of glucokinase mutations in monogenic forms of hyperinsulinemia of infancy, hyperglycemia and neonatal diabetes. Anna maintains an active research program in monogenic forms of diabetes and how genetics can be used to assist diabetes diagnosis and treatment. Her lab has expertise in variant characterization for multiple genes involved in monogenic diabetes and she supports clinicians with the interpretation of variants of unknown significance from genetic testing. A major focus of her current work is focused on how deep mutational scanning and maps of variant effects for diabetes relevant genes can be incorporated into guidelines for variant interpretation for monogenic diabetes through her involvement in the Clin Gen Monogenic Diabetes Variant Expert Review Panel and the Atlas of Variant Effects (AVE) consortium. Recently she has co-led the working group on Precision Diagnostics in monogenic diabetes for the and the ADA/EASD Precision Medicines Initiative which has uncovered a number of gaps in our knowledge.

Anna's research is not limited to monogenic forms of diabetes she is also an active member of multiple international consortia for genetic discovery for type 2 diabetes including the Accelerated Medicines Partnership for Common Metabolic Disease (AMP-CMD) where she uses her expertise in islet biology, functional genomics and cell and molecular physiology to bridge the gap between genetic discovery and biological and clinical insight. One of her areas of interest is in how genetics can be used for stratified medicine. Dr Gloyn is involved in several efforts to integrate genetic data on diabetes heterogeneity into human islet research within the Human Islet Research Network (HIRN). She is currently responsible for the genetic characterization of human islet donors for both the Integrated Islet Distribution Program (IIDP) where she heads the Human Genotyping Initiative (HIGI) and the Human Pancreas Atlas Program (HPAP) where she is responsible for the genotyping all donors. She has developed tools and methods to make genetic data available to islet users on ancestry and genetic risk for type 1 and type 2 diabetes.

Anna is an active member of multiple internal genetic discovery efforts including: NIH/Pharma funded Accelerated Medicines Partnership, DIAGRAM (Diabetes Genetics Replication and Meta-analysis), MAGIC (Meta-analysis of Glucose and Insulin traits Consortium), Type 2 Diabetes Genetic Exploration by Next-generation sequencing in multi-Ethnic Samples (T2D-GENES) and the Genetics of Type 2 Diabetes (GoT2D). She was also involved in the IMI funded STEMBANCC project which focused on delivering human IPS cell derived beta-cell models for drug discovery efforts.

2023-24 Courses


Stanford Advisees


Graduate and Fellowship Programs


All Publications


  • Islet autoantibodies as precision diagnostic tools to characterize heterogeneity in type 1 diabetes: a systematic review COMMUNICATIONS MEDICINE Felton, J. L., Redondo, M. J., Oram, R. A., Speake, C., Long, S., Onengut-Gumuscu, S., Rich, S. S., Monaco, G. F., Harris-Kawano, A., Perez, D., Saeed, Z., Hoag, B., Jain, R., Evans-Molina, C., DiMeglio, L. A., Ismail, H. M., Dabelea, D., Johnson, R. K., Urazbayeva, M., Wentworth, J. M., Grif, K. J., Sims, E. K., Tobias, D. K., Merino, J., Ahmad, A., Aiken, C., Benham, J. L., Bodhini, D., Clark, A. L., Colclough, K., Corcoy, R., Cromer, S. J., Duan, D., Felton, J. L., Francis, E. C., Gillard, P., Gingras, V., Gaillard, R., Haider, E., Hughes, A., Ikle, J. M., Jacobsen, L. M., Kahkoska, A. R., Kettunen, J. T., Kreienkamp, R. J., Lim, L., Mannisto, J. E., Massey, R., Mclennan, N., Miller, R. G., Morieri, M., Most, J., Naylor, R. N., Ozkan, B., Patel, K., Pilla, S. J., Prystupa, K., Raghavan, S., Rooney, M. R., Schoen, M., Semnani-Azad, Z., Sevilla-Gonzalez, M., Svalastoga, P., Takele, W., Tam, C., Thuesen, A. B., Tosur, M., Wallace, A. S., Wang, C. C., Wong, J. J., Yamamoto, J. M., Young, K., Amouyal, C., Andersen, M. K., Bonham, M. P., Chen, M., Cheng, F., Chikowore, T., Chivers, S. C., Clemmensen, C., Dabelea, D., Dawed, A. Y., Deutsch, A. J., Dickens, L. T., DiMeglio, L. A., Dudenhoffer-Pfeifer, M., Evans-Molina, C., Fernandez-Balsells, M., Fitipaldi, H., Fitzpatrick, S. L., Gitelman, S. E., Goodarzi, M. O., Grieger, J. A., Guasch-Ferre, M., Habibi, N., Hansen, T., Huang, C., Harris-Kawano, A., Ismail, H. M., Hoag, B., Johnson, R. K., Jones, A. G., Koivula, R. W., Leong, A., Leung, G. W., Libman, I. M., Liu, K., Long, S., Lowe, W. L., Morton, R. W., Motala, A. A., Onengut-Gumuscu, S., Pankow, J. S., Pathirana, M., Pazmino, S., Perez, D., Petrie, J. R., Powe, C. E., Quinteros, A., Jain, R., Ray, D., Ried-Larsen, M., Saeed, Z., Santhakumar, V., Kanbour, S., Sarkar, S., Monaco, G. F., Scholtens, D. M., Selvin, E., Sheu, W., Speake, C., Stanislawski, M. A., Steenackers, N., Steck, A. K., Stefan, N., Stoy, J., Taylor, R., Tye, S., Ukke, G., Urazbayeva, M., Van der Schueren, B., Vatier, C., Wentworth, J. M., Hannah, W., White, S. L., Yu, G., Zhang, Y., Zhou, S. J., Beltrand, J., Polak, M., Aukrust, I., de Franco, E., Flanagan, S. E., Maloney, K. A., McGovern, A., Molnes, J., Nakabuye, M., Njolstad, P., Pomares-Millan, H., Provenzano, M., Saint-Martin, C., Zhang, C., Zhu, Y., Auh, S., de Souza, R., Fawcett, A. J., Gruber, C., Mekonnen, E., Mixter, E., Sherifali, D., Eckel, R. H., Nolan, J. J., Philipson, L. H., Brown, R. J., Billings, L. K., Boyle, K., Costacou, T., Dennis, J. M., Florez, J. C., Gloyn, A. L., Gomez, M. F., Gottlieb, P. A., Greeley, S. W., Griffin, K., Hattersley, A. T., Hirsch, I. B., Hivert, M., Hood, K. K., Josefson, J. L., Kwak, S., Laffel, L. M., Lim, S. S., Loos, R. F., Ma, R. W., Mathieu, C., Mathioudakis, N., Meigs, J. B., Misra, S., Mohan, V., Murphy, R., Oram, R., Owen, K. R., Ozanne, S. E., Pearson, E. R., Perng, W., Pollin, T. I., Pop-Busui, R., Pratley, R. E., Redman, L. M., Redondo, M. J., Reynolds, R. M., Semple, R. K., Sherr, J. L., Sims, E. K., Sweeting, A., Tuomi, T., Udler, M. S., Vesco, K. K., Vilsboll, T., Wagner, R., Rich, S. S., Franks, P. W., ADA EASD PMDI 2024; 4 (1): 66

    Abstract

    Islet autoantibodies form the foundation for type 1 diabetes (T1D) diagnosis and staging, but heterogeneity exists in T1D development and presentation. We hypothesized that autoantibodies can identify heterogeneity before, at, and after T1D diagnosis, and in response to disease-modifying therapies.We systematically reviewed PubMed and EMBASE databases (6/14/2022) assessing 10 years of original research examining relationships between autoantibodies and heterogeneity before, at, after diagnosis, and in response to disease-modifying therapies in individuals at-risk or within 1 year of T1D diagnosis. A critical appraisal checklist tool for cohort studies was modified and used for risk of bias assessment.Here we show that 152 studies that met extraction criteria most commonly characterized heterogeneity before diagnosis (91/152). Autoantibody type/target was most frequently examined, followed by autoantibody number. Recurring themes included correlations of autoantibody number, type, and titers with progression, differing phenotypes based on order of autoantibody seroconversion, and interactions with age and genetics. Only 44% specifically described autoantibody assay standardization program participation.Current evidence most strongly supports the application of autoantibody features to more precisely define T1D before diagnosis. Our findings support continued use of pre-clinical staging paradigms based on autoantibody number and suggest that additional autoantibody features, particularly in relation to age and genetic risk, could offer more precise stratification. To improve reproducibility and applicability of autoantibody-based precision medicine in T1D, we propose a methods checklist for islet autoantibody-based manuscripts which includes use of precision medicine MeSH terms and participation in autoantibody standardization workshops.

    View details for DOI 10.1038/s43856-024-00478-y

    View details for Web of Science ID 001198030600001

    View details for PubMedID 38582818

    View details for PubMedCentralID PMC10998887

  • Multiplexed CRISPR gene editing in primary human islet cells with Cas9 ribonucleoprotein. iScience Bevacqua, R. J., Zhao, W., Merheb, E., Kim, S. H., Marson, A., Gloyn, A. L., Kim, S. K. 2024; 27 (1): 108693

    Abstract

    Successful genome editing in primary human islets could reveal features of the genetic regulatory landscape underlying beta cell function and diabetes risk. Here, we describe a CRISPR-based strategy to interrogate functions of predicted regulatory DNA elements using electroporation of a complex of Cas9 ribonucleoprotein (Cas9 RNP) and guide RNAs into primary human islet cells. We successfully targeted coding regions including the PDX1 exon 1, and non-coding DNA linked to diabetes susceptibility. CRISPR-Cas9 RNP approaches revealed genetic targets of regulation by DNA elements containing candidate diabetes risk SNPs, including an invivo enhancer of the MPHOSPH9 gene. CRISPR-Cas9 RNP multiplexed targeting of two cis-regulatory elements linked to diabetes risk in PCSK1, which encodes an endoprotease crucial for Insulin processing, also demonstrated efficient simultaneous editing of PCSK1 regulatory elements, resulting in impaired beta cell PCSK1 regulation and Insulin secretion. Multiplex CRISPR-Cas9 RNP provides powerful approaches to investigate and elucidate human islet cell gene regulation in health and diabetes.

    View details for DOI 10.1016/j.isci.2023.108693

    View details for PubMedID 38205242

  • Heterogeneity of increased biological age in type 2 diabetes correlates with differential tissue DNA methylation, biological variables, and pharmacological treatments. GeroScience Cortez, B. N., Pan, H., Hinthorn, S., Sun, H., Neretti, N., Gloyn, A. L., Aguayo-Mazzucato, C. 2023

    Abstract

    Biological age (BA) closely depicts age-related changes at a cellular level. Type 2 diabetes mellitus (T2D) accelerates BA when calculated using clinical biomarkers, but there is a large spread in the magnitude of individuals' age acceleration in T2D suggesting additional factors contributing to BA. Additionally, it is unknown whether BA can be changed with treatment. We hypothesized that potential determinants of the heterogeneous BA distribution in T2D could be due to differential tissue aging as reflected at the DNA methylation (DNAm) level, or biological variables and their respective therapeutic treatments. Publicly available DNAm samples were obtained to calculate BA using the DNAm phenotypic age (DNAmPhenoAge) algorithm. DNAmPhenoAge showed age acceleration in T2D samples of whole blood, pancreatic islets, and liver, but not in adipose tissue or skeletal muscle. Analysis of genes associated with differentially methylated CpG sites found a significant correlation between eight individual CpG methylation sites and gene expression. Clinical biomarkers from participants in the NHANES 2017-2018 and ACCORD cohorts were used to calculate BA using the Klemera and Doubal (KDM) method. Cardiovascular and glycemic biomarkers associated with increased BA while intensive blood pressure and glycemic management reduced BA to CA levels, demonstrating that accelerated BA can be restored in the setting of T2D.

    View details for DOI 10.1007/s11357-023-01009-8

    View details for PubMedID 37987887

  • Second international consensus report on gaps and opportunities for the clinical translation of precision diabetes medicine. Nature medicine Tobias, D. K., Merino, J., Ahmad, A., Aiken, C., Benham, J. L., Bodhini, D., Clark, A. L., Colclough, K., Corcoy, R., Cromer, S. J., Duan, D., Felton, J. L., Francis, E. C., Gillard, P., Gingras, V., Gaillard, R., Haider, E., Hughes, A., Ikle, J. M., Jacobsen, L. M., Kahkoska, A. R., Kettunen, J. L., Kreienkamp, R. J., Lim, L. L., Männistö, J. M., Massey, R., Mclennan, N. M., Miller, R. G., Morieri, M. L., Most, J., Naylor, R. N., Ozkan, B., Patel, K. A., Pilla, S. J., Prystupa, K., Raghavan, S., Rooney, M. R., Schön, M., Semnani-Azad, Z., Sevilla-Gonzalez, M., Svalastoga, P., Takele, W. W., Tam, C. H., Thuesen, A. C., Tosur, M., Wallace, A. S., Wang, C. C., Wong, J. J., Yamamoto, J. M., Young, K., Amouyal, C., Andersen, M. K., Bonham, M. P., Chen, M., Cheng, F., Chikowore, T., Chivers, S. C., Clemmensen, C., Dabelea, D., Dawed, A. Y., Deutsch, A. J., Dickens, L. T., DiMeglio, L. A., Dudenhöffer-Pfeifer, M., Evans-Molina, C., Fernández-Balsells, M. M., Fitipaldi, H., Fitzpatrick, S. L., Gitelman, S. E., Goodarzi, M. O., Grieger, J. A., Guasch-Ferré, M., Habibi, N., Hansen, T., Huang, C., Harris-Kawano, A., Ismail, H. M., Hoag, B., Johnson, R. K., Jones, A. G., Koivula, R. W., Leong, A., Leung, G. K., Libman, I. M., Liu, K., Long, S. A., Lowe, W. L., Morton, R. W., Motala, A. A., Onengut-Gumuscu, S., Pankow, J. S., Pathirana, M., Pazmino, S., Perez, D., Petrie, J. R., Powe, C. E., Quinteros, A., Jain, R., Ray, D., Ried-Larsen, M., Saeed, Z., Santhakumar, V., Kanbour, S., Sarkar, S., Monaco, G. S., Scholtens, D. M., Selvin, E., Sheu, W. H., Speake, C., Stanislawski, M. A., Steenackers, N., Steck, A. K., Stefan, N., Støy, J., Taylor, R., Tye, S. C., Ukke, G. G., Urazbayeva, M., Van der Schueren, B., Vatier, C., Wentworth, J. M., Hannah, W., White, S. L., Yu, G., Zhang, Y., Zhou, S. J., Beltrand, J., Polak, M., Aukrust, I., de Franco, E., Flanagan, S. E., Maloney, K. A., McGovern, A., Molnes, J., Nakabuye, M., Njølstad, P. R., Pomares-Millan, H., Provenzano, M., Saint-Martin, C., Zhang, C., Zhu, Y., Auh, S., de Souza, R., Fawcett, A. J., Gruber, C., Mekonnen, E. G., Mixter, E., Sherifali, D., Eckel, R. H., Nolan, J. J., Philipson, L. H., Brown, R. J., Billings, L. K., Boyle, K., Costacou, T., Dennis, J. M., Florez, J. C., Gloyn, A. L., Gomez, M. F., Gottlieb, P. A., Greeley, S. A., Griffin, K., Hattersley, A. T., Hirsch, I. B., Hivert, M. F., Hood, K. K., Josefson, J. L., Kwak, S. H., Laffel, L. M., Lim, S. S., Loos, R. J., Ma, R. C., Mathieu, C., Mathioudakis, N., Meigs, J. B., Misra, S., Mohan, V., Murphy, R., Oram, R., Owen, K. R., Ozanne, S. E., Pearson, E. R., Perng, W., Pollin, T. I., Pop-Busui, R., Pratley, R. E., Redman, L. M., Redondo, M. J., Reynolds, R. M., Semple, R. K., Sherr, J. L., Sims, E. K., Sweeting, A., Tuomi, T., Udler, M. S., Vesco, K. K., Vilsbøll, T., Wagner, R., Rich, S. S., Franks, P. W. 2023

    Abstract

    Precision medicine is part of the logical evolution of contemporary evidence-based medicine that seeks to reduce errors and optimize outcomes when making medical decisions and health recommendations. Diabetes affects hundreds of millions of people worldwide, many of whom will develop life-threatening complications and die prematurely. Precision medicine can potentially address this enormous problem by accounting for heterogeneity in the etiology, clinical presentation and pathogenesis of common forms of diabetes and risks of complications. This second international consensus report on precision diabetes medicine summarizes the findings from a systematic evidence review across the key pillars of precision medicine (prevention, diagnosis, treatment, prognosis) in four recognized forms of diabetes (monogenic, gestational, type 1, type 2). These reviews address key questions about the translation of precision medicine research into practice. Although not complete, owing to the vast literature on this topic, they revealed opportunities for the immediate or near-term clinical implementation of precision diabetes medicine; furthermore, we expose important gaps in knowledge, focusing on the need to obtain new clinically relevant evidence. Gaps include the need for common standards for clinical readiness, including consideration of cost-effectiveness, health equity, predictive accuracy, liability and accessibility. Key milestones are outlined for the broad clinical implementation of precision diabetes medicine.

    View details for DOI 10.1038/s41591-023-02502-5

    View details for PubMedID 37794253

    View details for PubMedCentralID 8563635

  • The use of precision diagnostics for monogenic diabetes: a systematic review and expert opinion. Communications medicine Murphy, R., Colclough, K., Pollin, T. I., Ikle, J. M., Svalastoga, P., Maloney, K. A., Saint-Martin, C., Molnes, J., Misra, S., Aukrust, I., de Franco, E., Flanagan, S. E., Njølstad, P. R., Billings, L. K., Owen, K. R., Gloyn, A. L. 2023; 3 (1): 136

    Abstract

    Monogenic diabetes presents opportunities for precision medicine but is underdiagnosed. This review systematically assessed the evidence for (1) clinical criteria and (2) methods for genetic testing for monogenic diabetes, summarized resources for (3) considering a gene or (4) variant as causal for monogenic diabetes, provided expert recommendations for (5) reporting of results; and reviewed (6) next steps after monogenic diabetes diagnosis and (7) challenges in precision medicine field.Pubmed and Embase databases were searched (1990-2022) using inclusion/exclusion criteria for studies that sequenced one or more monogenic diabetes genes in at least 100 probands (Question 1), evaluated a non-obsolete genetic testing method to diagnose monogenic diabetes (Question 2). The risk of bias was assessed using the revised QUADAS-2 tool. Existing guidelines were summarized for questions 3-5, and review of studies for questions 6-7, supplemented by expert recommendations. Results were summarized in tables and informed recommendations for clinical practice.There are 100, 32, 36, and 14 studies included for questions 1, 2, 6, and 7 respectively. On this basis, four recommendations for who to test and five on how to test for monogenic diabetes are provided. Existing guidelines for variant curation and gene-disease validity curation are summarized. Reporting by gene names is recommended as an alternative to the term MODY. Key steps after making a genetic diagnosis and major gaps in our current knowledge are highlighted.We provide a synthesis of current evidence and expert opinion on how to use precision diagnostics to identify individuals with monogenic diabetes.

    View details for DOI 10.1038/s43856-023-00369-8

    View details for PubMedID 37794142

    View details for PubMedCentralID 6058077

  • PAX4 loss of function increases diabetes risk by altering human pancreatic endocrine cell development. Nature communications Lau, H. H., Krentz, N. A., Abaitua, F., Perez-Alcantara, M., Chan, J. W., Ajeian, J., Ghosh, S., Lee, Y., Yang, J., Thaman, S., Champon, B., Sun, H., Jha, A., Hoon, S., Tan, N. S., Gardner, D. S., Kao, S. L., Tai, E. S., Gloyn, A. L., Teo, A. K. 2023; 14 (1): 6119

    Abstract

    The coding variant (p.Arg192His) in the transcription factor PAX4 is associated with an altered risk for type 2 diabetes (T2D) in East Asian populations. In mice, Pax4 is essential for beta cell formation but its role on human beta cell development and/or function is unknown. Participants carrying the PAX4 p.His192 allele exhibited decreased pancreatic beta cell function compared to homozygotes for the p.192Arg allele in a cross-sectional study in which we carried out an intravenous glucose tolerance test and an oral glucose tolerance test. In a pedigree of a patient with young onset diabetes, several members carry a newly identified p.Tyr186X allele. In the human beta cell model, EndoC-βH1, PAX4 knockdown led to impaired insulin secretion, reduced total insulin content, and altered hormone gene expression. Deletion of PAX4 in human induced pluripotent stem cell (hiPSC)-derived islet-like cells resulted in derepression of alpha cell gene expression. In vitro differentiation of hiPSCs carrying PAX4 p.His192 and p.X186 risk alleles exhibited increased polyhormonal endocrine cell formation and reduced insulin content that can be reversed with gene correction. Together, we demonstrate the role of PAX4 in human endocrine cell development, beta cell function, and its contribution to T2D-risk.

    View details for DOI 10.1038/s41467-023-41860-z

    View details for PubMedID 37777536

    View details for PubMedCentralID 5034897

  • Safe use of the ketogenic diet in an infant with microcephaly, epilepsy, and diabetes syndrome: a case report. BMC pediatrics Zegarra, W. A., Gallentine, W. B., Ruzhnikov, M. R., McAndrews, C. A., Gloyn, A. L., Addala, A. 2023; 23 (1): 453

    Abstract

    BACKGROUND: Microcephaly, epilepsy, and diabetes syndrome (MEDS) is a rare syndromic form of monogenic diabetes caused by bi-allelic loss of function mutations in IER3IP1. In vitro studies have shown that loss of IER31P leads to apoptosis in both neurons and pancreatic beta-cells. Simultaneous management of seizures and diabetes is challenging in patients with MEDS. We present the challenges and successes in the use of ketogenic diet in an infant with insulinopenic diabetes.CASE PRESENTATION: Our term female proband presented at 2 months of age with new onset multifocal seizures followed by the onset of infantile spasms (IS) at 4 months of age. An epilepsy gene panel identified bi-allelic variants, c.239T>G (p.Leu80*) and c.2T>A (initiator codon), in IER3IP1 that were subsequently shown to be inherited in trans. Following initiation of steroid therapy for IS, the patient developed clinically apparent insulin requiring diabetes. Her epilepsy was ultimately refractory to multiple antiseizure medications, thus the ketogenic diet (KD) was initiated. We were able to successfully titrate to a therapeutic KD ratio of 3:1 and maintain a ketotic state without diabetic ketoacidosis (DKA). With intercurrent illnesses, however, the patient had rapid decompensation and mild DKA due to delays in treatment, and for this reason, KD was discontinued after 5 months.CONCLUSIONS: We report two novel IER31P1 mutations in a patient with MEDS and the successful management of the cooccurring conditions of IS and insulinopenic diabetes with the KD. Our experience underscores the importance of careful monitoring during KD as our patient had DKA more easily when on the KD.

    View details for DOI 10.1186/s12887-023-04272-y

    View details for PubMedID 37689631

  • Management of Neonatal Diabetes due to a KCNJ11 Mutation with Automated Insulin Delivery System and Remote Patient Monitoring. Case reports in endocrinology Lee, M. Y., Gloyn, A. L., Maahs, D. M., Prahalad, P. 2023; 2023: 8825724

    Abstract

    Neonatal diabetes mellitus (NDM) is a monogenic form of diabetes. Management of hyperglycemia in neonates with subcutaneous insulin is challenging because of frequent feeding, variable quantity of milk intake with each feed, low insulin dose requirements, and high risk for hypoglycemia and associated complications in this population. We present a case of NDM in a proband initially presenting with focal seizures and diabetic ketoacidosis due to a pathologic mutation in the beta cell potassium ATP channel gene KCNJ11 c.679G > A (p.E227K). We describe the use of continuous glucose monitoring (CGM), insulin pump, automated insulin delivery system, and remote patient monitoring technologies to facilitate rapid and safe outpatient cross-titration from insulin to oral sulfonylurea. Our case highlights the safety and efficacy of these technologies for infants with diabetes, including improvements in glycemia, quality of life, and cost-effectiveness by shortening hospital stay.

    View details for DOI 10.1155/2023/8825724

    View details for PubMedID 37664823

    View details for PubMedCentralID PMC10468271

  • An Atlas of Variant Effects to understand the genome at nucleotide resolution. Genome biology Fowler, D. M., Adams, D. J., Gloyn, A. L., Hahn, W. C., Marks, D. S., Muffley, L. A., Neal, J. T., Roth, F. P., Rubin, A. F., Starita, L. M., Hurles, M. E. 2023; 24 (1): 147

    Abstract

    Sequencing has revealed hundreds of millions of human genetic variants, and continued efforts will only add to this variant avalanche. Insufficient information exists to interpret the effects of most variants, limiting opportunities for precision medicine and comprehension of genome function. A solution lies in experimental assessment of the functional effect of variants, which can reveal their biological and clinical impact. However, variant effect assays have generally been undertaken reactively for individual variants only after and, in most cases long after, their first observation. Now, multiplexed assays of variant effect can characterise massive numbers of variants simultaneously, yielding variant effect maps that reveal the function of every possible single nucleotide change in a gene or regulatory element. Generating maps for every protein encoding gene and regulatory element in the human genome would create an 'Atlas' of variant effect maps and transform our understanding of genetics and usher in a new era of nucleotide-resolution functional knowledge of the genome. An Atlas would reveal the fundamental biology of the human genome, inform human evolution, empower the development and use of therapeutics and maximize the utility of genomics for diagnosing and treating disease. The Atlas of Variant Effects Alliance is an international collaborative group comprising hundreds of researchers, technologists and clinicians dedicated to realising an Atlas of Variant Effects to help deliver on the promise of genomics.

    View details for DOI 10.1186/s13059-023-02986-x

    View details for PubMedID 37394429

    View details for PubMedCentralID PMC10316620

  • A comprehensive map of human glucokinase variant activity. Genome biology Gersing, S., Cagiada, M., Gebbia, M., Gjesing, A. P., Coté, A. G., Seesankar, G., Li, R., Tabet, D., Weile, J., Stein, A., Gloyn, A. L., Hansen, T., Roth, F. P., Lindorff-Larsen, K., Hartmann-Petersen, R. 2023; 24 (1): 97

    Abstract

    Glucokinase (GCK) regulates insulin secretion to maintain appropriate blood glucose levels. Sequence variants can alter GCK activity to cause hyperinsulinemic hypoglycemia or hyperglycemia associated with GCK-maturity-onset diabetes of the young (GCK-MODY), collectively affecting up to 10 million people worldwide. Patients with GCK-MODY are frequently misdiagnosed and treated unnecessarily. Genetic testing can prevent this but is hampered by the challenge of interpreting novel missense variants.Here, we exploit a multiplexed yeast complementation assay to measure both hyper- and hypoactive GCK variation, capturing 97% of all possible missense and nonsense variants. Activity scores correlate with in vitro catalytic efficiency, fasting glucose levels in carriers of GCK variants and with evolutionary conservation. Hypoactive variants are concentrated at buried positions, near the active site, and at a region of known importance for GCK conformational dynamics. Some hyperactive variants shift the conformational equilibrium towards the active state through a relative destabilization of the inactive conformation.Our comprehensive assessment of GCK variant activity promises to facilitate variant interpretation and diagnosis, expand our mechanistic understanding of hyperactive variants, and inform development of therapeutics targeting GCK.

    View details for DOI 10.1186/s13059-023-02935-8

    View details for PubMedID 37101203

    View details for PubMedCentralID PMC10131484

  • Small but mighty: microexons in glucose homeostasis. Trends in genetics : TIG Garcia, K., Gloyn, A. L. 2023

    Abstract

    Many molecular mechanisms underlying blood glucose homeostasis remain elusive. Juan-Mateu et al. find that pancreatic islet cells utilize a regulatory program, originally identified in neurons, that involves alternative splicing of microexons in genes important for insulin secretion or diabetes risk.

    View details for DOI 10.1016/j.tig.2023.04.003

    View details for PubMedID 37080883

  • Type 2 Diabetes risk alleles in Peptidyl-glycine Alpha-amidating Monooxygenase influence GLP-1 levels and response to GLP-1 Receptor Agonists. medRxiv : the preprint server for health sciences Umapathysivam, M. M., Araldi, E., Hastoy, B., Dawed, A. Y., Vatandaslar, H., Sengupta, S., Kaufmann, A., Thomsen, S., Hartmann, B., Jonsson, A. E., Kabakci, H., Thaman, S., Grarup, N., Have, C. T., Farch, K., Gjesing, A. P., Nawaz, S., Cheeseman, J., Neville, M. J., Pedersen, O., Walker, M., Jennison, C., Hattersley, A. T., Hansen, T., Karpe, F., Holst, J. J., Jones, A. G., Ristow, M., McCarthy, M. I., Pearson, E. R., Stoffel, M., Gloyn, A. L. 2023

    Abstract

    Patients with type 2 diabetes vary in their response to currently available therapeutic agents (including GLP-1 receptor agonists) leading to suboptimal glycemic control and increased risk of complications. We show that human carriers of hypomorphic T2D-risk alleles in the gene encoding peptidyl-glycine alpha-amidating monooxygenase (PAM), as well as Pam- knockout mice, display increased resistance to GLP-1 in vivo . Pam inactivation in mice leads to reduced gastric GLP-1R expression and faster gastric emptying: this persists during GLP-1R agonist treatment and is rescued when GLP-1R activity is antagonized, indicating resistance to GLP-1's gastric slowing properties. Meta-analysis of human data from studies examining GLP-1R agonist response (including RCTs) reveals a relative loss of 44% and 20% of glucose lowering (measured by glycated hemoglobin) in individuals with hypomorphic PAM alleles p.S539W and p.D536G treated with GLP-1R agonist. Genetic variation in PAM has effects on incretin signaling that alters response to medication used commonly for treatment of T2D. (Funded by the Wellcome, Medical Research Council, European Union, NIHR Oxford Biomedical Research Centre, United Kingdom, Registered on ClinicalTrials.gov, NCT02723110 .).Summary Paragraph: Type 2 diabetes (T2D) is a leading cause of morbidity and mortality globally 1 . Current management of T2D patients focuses on lowering glycemic exposure and reducing complications with lifestyle and pharmacological interventions 2 . Despite the availability of multiple medications to lower glycated hemoglobin (HbA1c), only 53% of individuals with T2D reach the glycemic target (HbA1c <7%) 3, 4 . There is potential to improve medication selection through "precision medicine" where patient specific factors (e.g. genetic markers) are used to indicate whether a patient is more or less likely to respond to a medication. Here we show that human carriers of hypomorphic T2D-risk alleles in the gene encoding peptidyl-glycine alpha-amidating monooxygenase (PAM), as well as Pam- knockout mice, have reduced PAM enzyme activity, display increased resistance to glucagon like peptide 1 (GLP-1) in vivo and have reduced response to the GLP-1 receptor agonist. Meta-analysis of human data from studies examining GLP-1 receptor agonist response (including RCTs) reveals a relative loss of 44% and 20% of glucose lowering (measured by glycated hemoglobin) in individuals with hypomorphic PAM alleles p.S539W and p.D536G treated with GLP-1 receptor agonist. Genetic variation in PAM has effects on incretin signaling that alters response to medication used commonly for treatment of T2D.

    View details for DOI 10.1101/2023.04.07.23288197

    View details for PubMedID 37090505

  • The Type 2 Diabetes Knowledge Portal: An open access genetic resource dedicated to type 2 diabetes and related traits. Cell metabolism Costanzo, M. C., von Grotthuss, M., Massung, J., Jang, D., Caulkins, L., Koesterer, R., Gilbert, C., Welch, R. P., Kudtarkar, P., Hoang, Q., Boughton, A. P., Singh, P., Sun, Y., Duby, M., Moriondo, A., Nguyen, T., Smadbeck, P., Alexander, B. R., Brandes, M., Carmichael, M., Dornbos, P., Green, T., Huellas-Bruskiewicz, K. C., Ji, Y., Kluge, A., McMahon, A. C., Mercader, J. M., Ruebenacker, O., Sengupta, S., Spalding, D., Taliun, D., AMP-T2D Consortium, Smith, P., Thomas, M. K., Akolkar, B., Brosnan, M. J., Cherkas, A., Chu, A. Y., Fauman, E. B., Fox, C. S., Kamphaus, T. N., Miller, M. R., Nguyen, L., Parsa, A., Reilly, D. F., Ruetten, H., Wholley, D., Zaghloul, N. A., Abecasis, G. R., Altshuler, D., Keane, T. M., McCarthy, M. I., Gaulton, K. J., Florez, J. C., Boehnke, M., Burtt, N. P., Flannick, J., Abecasis, G., Akolkar, B., Alexander, B. R., Allred, N. D., Altshuler, D., Below, J. E., Bergman, R., Beulens, J. W., Blangero, J., Boehnke, M., Bokvist, K., Bottinger, E., Boughton, A. P., Bowden, D., Brosnan, M. J., Brown, C., Bruskiewicz, K., Burtt, N. P., Carmichael, M., Caulkins, L., Cebola, I., Chambers, J., Ida Chen, Y., Cherkas, A., Chu, A. Y., Clark, C., Claussnitzer, M., Costanzo, M. C., Cox, N. J., Hoed, M. d., Dong, D., Duby, M., Duggirala, R., Dupuis, J., Elders, P. J., Engreitz, J. M., Fauman, E., Ferrer, J., Flannick, J., Flicek, P., Flickinger, M., Florez, J. C., Fox, C. S., Frayling, T. M., Frazer, K. A., Gaulton, K. J., Gilbert, C., Gloyn, A. L., Green, T., Hanis, C. L., Hanson, R., Hattersley, A. T., Hoang, Q., Im, H. K., Iqbal, S., Jacobs, S. B., Jang, D., Jordan, T., Kamphaus, T., Karpe, F., Keane, T. M., Kim, S. K., Kluge, A., Koesterer, R., Kudtarkar, P., Lage, K., Lange, L. A., Lazar, M., Lehman, D., Liu, C., Loos, R. J., Ma, R. C., MacDonald, P., Massung, J., Maurano, M. T., McCarthy, M. I., McVean, G., Meigs, J. B., Mercader, J. M., Miller, M. R., Mitchell, B., Mohlke, K. L., Morabito, S., Morgan, C., Mullican, S., Narendra, S., Ng, M. C., Nguyen, L., Palmer, C. N., Parker, S. C., Parrado, A., Parsa, A., Pawlyk, A. C., Pearson, E. R., Plump, A., Province, M., Quertermous, T., Redline, S., Reilly, D. F., Ren, B., Rich, S. S., Richards, J. B., Rotter, J. I., Ruebenacker, O., Ruetten, H., Salem, R. M., Sander, M., Sanders, M., Sanghera, D., Scott, L. J., Sengupta, S., Siedzik, D., Sim, X., Singh, P., Sladek, R., Small, K., Smith, P., Stein, P., Spalding, D., Stringham, H. M., Sun, Y., Susztak, K., 't Hart, L. M., Taliun, D., Taylor, K., Thomas, M. K., Todd, J. A., Udler, M. S., Voight, B., von Grotthuss, M., Wan, A., Welch, R. P., Wholley, D., Yuksel, K., Zaghloul, N. A. 2023

    Abstract

    Associations between human genetic variation and clinical phenotypes have become a foundation of biomedical research. Most repositories of these data seek to be disease-agnostic and therefore lack disease-focused views. The Type 2 Diabetes Knowledge Portal (T2DKP) is a public resource of genetic datasets and genomic annotations dedicated to type 2 diabetes (T2D) and related traits. Here, we seek to make the T2DKP more accessible to prospective users and more useful to existing users. First, we evaluate the T2DKP's comprehensiveness by comparing its datasets with those of other repositories. Second, we describe how researchers unfamiliar with human genetic data can begin using and correctly interpreting them via the T2DKP. Third, we describe how existing users can extend their current workflows to use the full suite of tools offered by the T2DKP. We finally discuss the lessons offered by the T2DKP toward the goal of democratizing access to complex disease genetic results.

    View details for DOI 10.1016/j.cmet.2023.03.001

    View details for PubMedID 36963395

  • Loss of RREB1 in pancreatic beta cells reduces cellular insulin content and affects endocrine cell gene expression. Diabetologia Mattis, K. K., Krentz, N. A., Metzendorf, C., Abaitua, F., Spigelman, A. F., Sun, H., Ikle, J. M., Thaman, S., Rottner, A. K., Bautista, A., Mazzaferro, E., Perez-Alcantara, M., Manning Fox, J. E., Torres, J. M., Wesolowska-Andersen, A., Yu, G. Z., Mahajan, A., Larsson, A., MacDonald, P. E., Davies, B., den Hoed, M., Gloyn, A. L. 2023

    Abstract

    Genome-wide studies have uncovered multiple independent signals at the RREB1 locus associated with altered type 2 diabetes risk and related glycaemic traits. However, little is known about the function of the zinc finger transcription factor Ras-responsive element binding protein 1 (RREB1) in glucose homeostasis or how changes in its expression and/or function influence diabetes risk.A zebrafish model lacking rreb1a and rreb1b was used to study the effect of RREB1 loss in vivo. Using transcriptomic and cellular phenotyping of a human beta cell model (EndoC-βH1) and human induced pluripotent stem cell (hiPSC)-derived beta-like cells, we investigated how loss of RREB1 expression and activity affects pancreatic endocrine cell development and function. Ex vivo measurements of human islet function were performed in donor islets from carriers of RREB1 type 2 diabetes risk alleles.CRISPR/Cas9-mediated loss of rreb1a and rreb1b function in zebrafish supports an in vivo role for the transcription factor in beta cell mass, beta cell insulin expression and glucose levels. Loss of RREB1 also reduced insulin gene expression and cellular insulin content in EndoC-βH1 cells and impaired insulin secretion under prolonged stimulation. Transcriptomic analysis of RREB1 knockdown and knockout EndoC-βH1 cells supports RREB1 as a novel regulator of genes involved in insulin secretion. In vitro differentiation of RREB1KO/KO hiPSCs revealed dysregulation of pro-endocrine cell genes, including RFX family members, suggesting that RREB1 also regulates genes involved in endocrine cell development. Human donor islets from carriers of type 2 diabetes risk alleles in RREB1 have altered glucose-stimulated insulin secretion ex vivo, consistent with a role for RREB1 in regulating islet cell function.Together, our results indicate that RREB1 regulates beta cell function by transcriptionally regulating the expression of genes involved in beta cell development and function.

    View details for DOI 10.1007/s00125-022-05856-6

    View details for PubMedID 36633628

  • Inferring causal genes at type 2 diabetes GWAS loci through chromosome interactions in islet cells. Wellcome open research Torres, J. M., Sun, H., Nylander, V., Downes, D. J., van de Bunt, M., McCarthy, M. I., Hughes, J. R., Gloyn, A. L. 2023; 8: 165

    Abstract

    Background: Resolving causal genes for type 2 diabetes at loci implicated by genome-wide association studies (GWAS) requires integrating functional genomic data from relevant cell types. Chromatin features in endocrine cells of the pancreatic islet are particularly informative and recent studies leveraging chromosome conformation capture (3C) with Hi-C based methods have elucidated regulatory mechanisms in human islets. However, these genome-wide approaches are less sensitive and afford lower resolution than methods that target specific loci. Methods: To gauge the extent to which targeted 3C further resolves chromatin-mediated regulatory mechanisms at GWAS loci, we generated interaction profiles at 23 loci using next-generation (NG) capture-C in a human beta cell model (EndoC-betaH1) and contrasted these maps with Hi-C maps in EndoC-betaH1 cells and human islets and a promoter capture Hi-C map in human islets. Results: We found improvements in assay sensitivity of up to 33-fold and resolved ~3.6X more chromatin interactions. At a subset of 18 loci with 25 co-localised GWAS and eQTL signals, NG Capture-C interactions implicated effector transcripts at five additional genetic signals relative to promoter capture Hi-C through physical contact with gene promoters. Conclusions: High resolution chromatin interaction profiles at selectively targeted loci can complement genome- and promoter-wide maps.

    View details for DOI 10.12688/wellcomeopenres.18653.2

    View details for PubMedID 37736013

  • A genome-wide CRISPR screen identifies CALCOCO2 as a regulator of beta cell function influencing type 2 diabetes risk. Nature genetics Rottner, A. K., Ye, Y., Navarro-Guerrero, E., Rajesh, V., Pollner, A., Bevacqua, R. J., Yang, J., Spigelman, A. F., Baronio, R., Bautista, A., Thomsen, S. K., Lyon, J., Nawaz, S., Smith, N., Wesolowska-Andersen, A., Fox, J. E., Sun, H., Kim, S. K., Ebner, D., MacDonald, P. E., Gloyn, A. L. 2022

    Abstract

    Identification of the genes and processes mediating genetic association signals for complex diseases represents a major challenge. As many of the genetic signals for type 2 diabetes (T2D) exert their effects through pancreatic islet-cell dysfunction, we performed a genome-wide pooled CRISPR loss-of-function screen in a human pancreatic beta cell line. We assessed the regulation of insulin content as a disease-relevant readout of beta cell function and identified 580 genes influencing this phenotype. Integration with genetic and genomic data provided experimental support for 20 candidate T2D effector transcripts including the autophagy receptor CALCOCO2. Loss of CALCOCO2 was associated with distorted mitochondria, less proinsulin-containing immature granules and accumulation of autophagosomes upon inhibition of late-stage autophagy. Carriers of T2D-associated variants at the CALCOCO2 locus further displayed altered insulin secretion. Our study highlights how cellular screens can augment existing multi-omic efforts to support mechanistic understanding and provide evidence for causal effects at genome-wide association studies loci.

    View details for DOI 10.1038/s41588-022-01261-2

    View details for PubMedID 36543916

  • Zmiz1 is required for mature β-cell function and mass expansion upon high fat feeding. Molecular metabolism Alghamdi, T. A., Krentz, N. A., Smith, N., Spigelman, A. F., Rajesh, V., Jha, A., Ferdaoussi, M., Suzuki, K., Yang, J., Manning Fox, J. E., Sun, H., Sun, Z., Gloyn, A. L., MacDonald, P. E. 2022: 101621

    Abstract

    Identifying the transcripts which mediate genetic association signals for type 2 diabetes (T2D) is critical to understand disease mechanisms. Studies in pancreatic islets support the transcription factor ZMIZ1 as a transcript underlying a T2D GWAS signal, but how it influences T2D risk is unknown.β-cell-specific Zmiz1 knockout (Zmiz1βKO) mice were generated and phenotypically characterised. Glucose homeostasis was assessed in Zmiz1βKO mice and their control littermates on chow diet (CD) and high fat diet (HFD). Islet morphology and function were examined by immunocytochemistry and in vitro islet function was assessed by dynamic insulin secretion assay. Transcript and protein expression were assessed by RNA sequencing and Western blotting. In islets isolated from genotyped human donors, we assessed glucose-dependent insulin secretion and islet insulin content by static incubation assay.Male and female Zmiz1βKO mice were glucose intolerant with impaired insulin secretion, compared with control littermates. Transcriptomic profiling of Zmiz1βKO islets identified over 500 differentially expressed genes including those involved in β-cell function and maturity, which we confirmed at the protein level. Upon HFD, Zmiz1βKO mice fail to expand β-cell mass and become severely diabetic. Human islets from carriers of the ZMIZ1-linked T2D-risk alleles have reduced islet insulin content and glucose-stimulated insulin secretion.β-cell Zmiz1 is required for normal glucose homeostasis. Genetic variation at the ZMIZ1 locus may influence T2D-risk by reducing islet mass expansion upon metabolic stress and the ability to maintain a mature β-cell state.

    View details for DOI 10.1016/j.molmet.2022.101621

    View details for PubMedID 36307047

  • The contribution of functional HNF1A variants and polygenic susceptibility to risk of type 2 diabetes in ancestrally diverse populations. Diabetologia Stalbow, L. A., Preuss, M. H., Smit, R. A., Chami, N., Bjørkhaug, L., Aukrust, I., Gloyn, A. L., Loos, R. J. 2022

    Abstract

    We examined the contribution of rare HNF1A variants to type 2 diabetes risk and age of diagnosis, and the extent to which their impact is affected by overall genetic susceptibility, across three ancestry groups.Using exome sequencing data of 160,615 individuals of the UK Biobank and 18,797 individuals of the BioMe Biobank, we identified 746 carriers of rare functional HNF1A variants (minor allele frequency ≤1%), of which 507 carry variants in the functional domains. We calculated polygenic risk scores (PRSs) based on genome-wide association study summary statistics for type 2 diabetes, and examined the association of HNF1A variants and PRS with risk of type 2 diabetes and age of diagnosis. We also tested whether the PRS affects the association between HNF1A variants and type 2 diabetes risk by including an interaction term.Rare HNF1A variants that are predicted to impair protein function are associated with increased risk of type 2 diabetes in individuals of European ancestry (OR 1.46, p=0.049), particularly when the variants are located in the functional domains (OR 1.89, p=0.002). No association was observed for individuals of African ancestry (OR 1.10, p=0.60) or Hispanic-Latino ancestry (OR 1.00, p=1.00). Rare functional HNF1A variants were associated with an earlier age at diagnosis in the Hispanic-Latino population (β=-5.0 years, p=0.03), and this association was marginally more pronounced for variants in the functional domains (β=-5.59 years, p=0.03). No associations were observed for other ancestries (African ancestry β=-2.7 years, p=0.13; European ancestry β=-3.5 years, p=0.20). A higher PRS was associated with increased odds of type 2 diabetes in all ancestries (OR 1.61-2.11, p<10-5) and an earlier age at diagnosis in individuals of African ancestry (β=-1.4 years, p=3.7 × 10-6) and Hispanic-Latino ancestry (β=-2.4 years, p<2 × 10-16). Furthermore, a higher PRS exacerbated the effect of the functional HNF1A variants on type 2 diabetes in the European ancestry population (pinteraction=0.037).We show that rare functional HNF1A variants, in particular those located in the functional domains, increase the risk of type 2 diabetes, at least among individuals of European ancestry. Their effect is even more pronounced in individuals with a high polygenic susceptibility. Our analyses highlight the importance of the location of functional variants within a gene and an individual's overall polygenic susceptibility, and emphasise the need for more genetic data in non-European populations.

    View details for DOI 10.1007/s00125-022-05806-2

    View details for PubMedID 36216889

  • Genetic regulation of RNA splicing in human pancreatic islets. Genome biology Atla, G., Bonas-Guarch, S., Cuenca-Ardura, M., Beucher, A., Crouch, D. J., Garcia-Hurtado, J., Moran, I., T2DSystems Consortium, Irimia, M., Prasad, R. B., Gloyn, A. L., Marselli, L., Suleiman, M., Berney, T., de Koning, E. J., Kerr-Conte, J., Pattou, F., Todd, J. A., Piemonti, L., Ferrer, J., Cnop, M., Eliasson, L., Esguerra, J. L., Eizirik, D. L., Groop, L., Jensen, T. S., Hansen, T., Marchetti, P., Mercader, J. M., Mulder, H., Stabile-Barnett, C. R., Thirion, C., Torrents, D. 2022; 23 (1): 196

    Abstract

    BACKGROUND: Non-coding genetic variants that influence gene transcription in pancreatic islets play a major role in the susceptibility to type 2 diabetes (T2D), and likely also contribute to type 1 diabetes (T1D) risk. For many loci, however, the mechanisms through which non-coding variants influence diabetes susceptibility are unknown.RESULTS: We examine splicing QTLs (sQTLs) in pancreatic islets from 399 human donors and observe that commongenetic variation has a widespread influence on thesplicing of genes with established roles in islet biology and diabetes. In parallel, we profile expression QTLs (eQTLs) and use transcriptome-wide association as well as genetic co-localization studies to assign islet sQTLs or eQTLs to T2D and T1D susceptibility signals, many of which lack candidate effector genes. This analysis reveals biologically plausible mechanisms, including the association of T2D with an sQTL that creates a nonsense isoform in ERO1B, a regulator of ER-stress and proinsulin biosynthesis. The expanded list of T2D risk effector genes reveals overrepresented pathways, including regulators of G-protein-mediated cAMP production. The analysis of sQTLs also reveals candidate effector genes for T1D susceptibility such as DCLRE1B, a senescence regulator, and lncRNA MEG3.CONCLUSIONS: These data expose widespread effects of common genetic variants on RNA splicing in pancreatic islets. The results support a role for splicing variation in diabetes susceptibility, and offera new set of genetic targets with potential therapeutic benefit.

    View details for DOI 10.1186/s13059-022-02757-0

    View details for PubMedID 36109769

  • Every islet matters: improving the impact of human islet research. Nature metabolism Gloyn, A. L., Ibberson, M., Marchetti, P., Powers, A. C., Rorsman, P., Sander, M., Solimena, M. 2022

    Abstract

    Detailed characterization of human pancreatic islets is key to elucidating the pathophysiology of all forms of diabetes, especially type 2 diabetes. However, access to human pancreatic islets is limited. Pancreatic tissue for islet retrieval can be obtained from brain-dead organ donors or from individuals undergoing pancreatectomy, often referred to as 'living donors'. Different protocols for human islet procurement can substantially impact islet function. This variability, coupled with heterogeneity between individuals and islets, results in analytical challenges to separate genuine disease pathology or differences between human donors from experimental noise. There are currently no international guidelines for human donor phenotyping, islet procurement and functional characterization. This lack of standardization means that substantial investments from multiple international efforts towards improved understanding of diabetes pathology cannot be fully leveraged. In this Perspective, we overview the status of the field of human islet research, highlight the challenges and propose actions that could accelerate research progress and increase understanding of type 2 diabetes to slow its pandemic spreading.

    View details for DOI 10.1038/s42255-022-00607-8

    View details for PubMedID 35953581

  • Multi-ancestry genetic study of type 2 diabetes highlights the power of diverse populations for discovery and translation. Nature genetics Mahajan, A., Spracklen, C. N., Zhang, W., Ng, M. C., Petty, L. E., Kitajima, H., Yu, G. Z., Rueger, S., Speidel, L., Kim, Y. J., Horikoshi, M., Mercader, J. M., Taliun, D., Moon, S., Kwak, S., Robertson, N. R., Rayner, N. W., Loh, M., Kim, B., Chiou, J., Miguel-Escalada, I., Della Briotta Parolo, P., Lin, K., Bragg, F., Preuss, M. H., Takeuchi, F., Nano, J., Guo, X., Lamri, A., Nakatochi, M., Scott, R. A., Lee, J., Huerta-Chagoya, A., Graff, M., Chai, J., Parra, E. J., Yao, J., Bielak, L. F., Tabara, Y., Hai, Y., Steinthorsdottir, V., Cook, J. P., Kals, M., Grarup, N., Schmidt, E. M., Pan, I., Sofer, T., Wuttke, M., Sarnowski, C., Gieger, C., Nousome, D., Trompet, S., Long, J., Sun, M., Tong, L., Chen, W., Ahmad, M., Noordam, R., Lim, V. J., Tam, C. H., Joo, Y. Y., Chen, C., Raffield, L. M., Lecoeur, C., Prins, B. P., Nicolas, A., Yanek, L. R., Chen, G., Jensen, R. A., Tajuddin, S., Kabagambe, E. K., An, P., Xiang, A. H., Choi, H. S., Cade, B. E., Tan, J., Flanagan, J., Abaitua, F., Adair, L. S., Adeyemo, A., Aguilar-Salinas, C. A., Akiyama, M., Anand, S. S., Bertoni, A., Bian, Z., Bork-Jensen, J., Brandslund, I., Brody, J. A., Brummett, C. M., Buchanan, T. A., Canouil, M., Chan, J. C., Chang, L., Chee, M., Chen, J., Chen, S., Chen, Y., Chen, Z., Chuang, L., Cushman, M., Das, S. K., de Silva, H. J., Dedoussis, G., Dimitrov, L., Doumatey, A. P., Du, S., Duan, Q., Eckardt, K., Emery, L. S., Evans, D. S., Evans, M. K., Fischer, K., Floyd, J. S., Ford, I., Fornage, M., Franco, O. H., Frayling, T. M., Freedman, B. I., Fuchsberger, C., Genter, P., Gerstein, H. C., Giedraitis, V., Gonzalez-Villalpando, C., Gonzalez-Villalpando, M. E., Goodarzi, M. O., Gordon-Larsen, P., Gorkin, D., Gross, M., Guo, Y., Hackinger, S., Han, S., Hattersley, A. T., Herder, C., Howard, A., Hsueh, W., Huang, M., Huang, W., Hung, Y., Hwang, M. Y., Hwu, C., Ichihara, S., Ikram, M. A., Ingelsson, M., Islam, M. T., Isono, M., Jang, H., Jasmine, F., Jiang, G., Jonas, J. B., Jorgensen, M. E., Jorgensen, T., Kamatani, Y., Kandeel, F. R., Kasturiratne, A., Katsuya, T., Kaur, V., Kawaguchi, T., Keaton, J. M., Kho, A. N., Khor, C., Kibriya, M. G., Kim, D., Kohara, K., Kriebel, J., Kronenberg, F., Kuusisto, J., Lall, K., Lange, L. A., Lee, M., Lee, N. R., Leong, A., Li, L., Li, Y., Li-Gao, R., Ligthart, S., Lindgren, C. M., Linneberg, A., Liu, C., Liu, J., Locke, A. E., Louie, T., Luan, J., Luk, A. O., Luo, X., Lv, J., Lyssenko, V., Mamakou, V., Mani, K. R., Meitinger, T., Metspalu, A., Morris, A. D., Nadkarni, G. N., Nadler, J. L., Nalls, M. A., Nayak, U., Nongmaithem, S. S., Ntalla, I., Okada, Y., Orozco, L., Patel, S. R., Pereira, M. A., Peters, A., Pirie, F. J., Porneala, B., Prasad, G., Preissl, S., Rasmussen-Torvik, L. J., Reiner, A. P., Roden, M., Rohde, R., Roll, K., Sabanayagam, C., Sander, M., Sandow, K., Sattar, N., Schonherr, S., Schurmann, C., Shahriar, M., Shi, J., Shin, D. M., Shriner, D., Smith, J. A., So, W. Y., Stancakova, A., Stilp, A. M., Strauch, K., Suzuki, K., Takahashi, A., Taylor, K. D., Thorand, B., Thorleifsson, G., Thorsteinsdottir, U., Tomlinson, B., Torres, J. M., Tsai, F., Tuomilehto, J., Tusie-Luna, T., Udler, M. S., Valladares-Salgado, A., van Dam, R. M., van Klinken, J. B., Varma, R., Vujkovic, M., Wacher-Rodarte, N., Wheeler, E., Whitsel, E. A., Wickremasinghe, A. R., van Dijk, K. W., Witte, D. R., Yajnik, C. S., Yamamoto, K., Yamauchi, T., Yengo, L., Yoon, K., Yu, C., Yuan, J., Yusuf, S., Zhang, L., Zheng, W., FinnGen, eMERGE Consortium, Raffel, L. J., Igase, M., Ipp, E., Redline, S., Cho, Y. S., Lind, L., Province, M. A., Hanis, C. L., Peyser, P. A., Ingelsson, E., Zonderman, A. B., Psaty, B. M., Wang, Y., Rotimi, C. N., Becker, D. M., Matsuda, F., Liu, Y., Zeggini, E., Yokota, M., Rich, S. S., Kooperberg, C., Pankow, J. S., Engert, J. C., Chen, Y. I., Froguel, P., Wilson, J. G., Sheu, W. H., Kardia, S. L., Wu, J., Hayes, M. G., Ma, R. C., Wong, T., Groop, L., Mook-Kanamori, D. O., Chandak, G. R., Collins, F. S., Bharadwaj, D., Pare, G., Sale, M. M., Ahsan, H., Motala, A. A., Shu, X., Park, K., Jukema, J. W., Cruz, M., McKean-Cowdin, R., Grallert, H., Cheng, C., Bottinger, E. P., Dehghan, A., Tai, E., Dupuis, J., Kato, N., Laakso, M., Kottgen, A., Koh, W., Palmer, C. N., Liu, S., Abecasis, G., Kooner, J. S., Loos, R. J., North, K. E., Haiman, C. A., Florez, J. C., Saleheen, D., Hansen, T., Pedersen, O., Magi, R., Langenberg, C., Wareham, N. J., Maeda, S., Kadowaki, T., Lee, J., Millwood, I. Y., Walters, R. G., Stefansson, K., Myers, S. R., Ferrer, J., Gaulton, K. J., Meigs, J. B., Mohlke, K. L., Gloyn, A. L., Bowden, D. W., Below, J. E., Chambers, J. C., Sim, X., Boehnke, M., Rotter, J. I., McCarthy, M. I., Morris, A. P., Rueger, S., Della Briotta Parolo, P., Joo, Y. Y., Hayes, M. G. 2022

    Abstract

    We assembled an ancestrally diverse collection of genome-wide association studies (GWAS) of type 2 diabetes (T2D) in 180,834 affected individuals and 1,159,055 controls (48.9% non-European descent) through the Diabetes Meta-Analysis of Trans-Ethnic association studies (DIAMANTE) Consortium. Multi-ancestry GWAS meta-analysis identified 237 loci attaining stringent genome-wide significance (P<5*10-9), which were delineated to 338 distinct association signals. Fine-mapping of these signals was enhanced by the increased sample size and expanded population diversity of the multi-ancestry meta-analysis, which localized 54.4% of T2D associations to a single variant with >50% posterior probability. This improved fine-mapping enabled systematic assessment of candidate causal genes and molecular mechanisms through which T2D associations are mediated, laying the foundations for functional investigations. Multi-ancestry genetic risk scores enhanced transferability of T2D prediction across diverse populations. Our study provides a step toward more effective clinical translation of T2D GWAS to improve global health for all, irrespective of genetic background.

    View details for DOI 10.1038/s41588-022-01058-3

    View details for PubMedID 35551307

  • TIGER: The gene expression regulatory variation landscape of human pancreatic islets. Cell reports Alonso, L., Piron, A., Moran, I., Guindo-Martinez, M., Bonas-Guarch, S., Atla, G., Miguel-Escalada, I., Royo, R., Puiggros, M., Garcia-Hurtado, X., Suleiman, M., Marselli, L., Esguerra, J. L., Turatsinze, J., Torres, J. M., Nylander, V., Chen, J., Eliasson, L., Defrance, M., Amela, R., MAGIC, Mulder, H., Gloyn, A. L., Groop, L., Marchetti, P., Eizirik, D. L., Ferrer, J., Mercader, J. M., Cnop, M., Torrents, D. 2021; 37 (2): 109807

    Abstract

    Genome-wide association studies (GWASs) identified hundreds of signals associated with type 2 diabetes (T2D). To gain insight into their underlying molecular mechanisms, we have created the translational human pancreatic islet genotype tissue-expression resource (TIGER), aggregating >500 human islet genomic datasets from five cohorts in the Horizon 2020 consortium T2DSystems. We impute genotypes using four reference panels and meta-analyze cohorts to improve the coverage of expression quantitative trait loci (eQTL) and develop a method to combine allele-specific expression across samples (cASE). We identify >1 million islet eQTLs, 53 of which colocalize with T2D signals. Among them, a low-frequency allele that reduces T2D risk by half increases CCND2 expression. We identify eight cASE colocalizations, among which we found a T2D-associated SLC30A8 variant. We make all data available through the TIGER portal (http://tiger.bsc.es), which represents a comprehensive human islet genomic data resource to elucidate how genetic variation affects islet function and translates into therapeutic insight and precision medicine for T2D.

    View details for DOI 10.1016/j.celrep.2021.109807

    View details for PubMedID 34644572

  • Genetics of Type 2 Diabetes: Opportunities for Precision Medicine: JACC Focus Seminar. Journal of the American College of Cardiology Kim, D. S., Gloyn, A. L., Knowles, J. W. 2021; 78 (5): 496-512

    Abstract

    Type 2 diabetes (T2D) is highly prevalent and is a strong contributor for cardiovascular disease. However, there is significant heterogeneity in disease pathogenesis and the risk of complications. Enormous progress has been made in our ability to catalog genetic variation associated with T2D risk and variation in disease-relevant quantitative traits. These discoveries hold the potential to shed light on tractable targets and pathways for safe and effective therapeutic development, but the promise of precision medicine has been slow to be realized. Recent studies have identified subgroups of individuals with differential risk for intermediate phenotypes (eg, lipid levels, fasting insulin, body mass index) that contribute to T2D risk, helping to account for the observed clinical heterogeneity. These "partitioned genetic risk scores" not only have the potential to identify patients at greatest risk of cardiovascular disease and rapid disease progression, but also could aid patient stratification bridging the gap toward precision medicine for T2D.

    View details for DOI 10.1016/j.jacc.2021.03.346

    View details for PubMedID 34325839

  • A brief history of diabetes genetics: insights for pancreatic beta-cell development and function. The Journal of endocrinology Ikle, J. M., Gloyn, A. L. 2021

    Abstract

    Since the discovery of insulin 100 years ago, our knowledge and understanding of diabetes has grown exponentially. Specifically, with regards to the genetics underlying diabetes risk, our discoveries have paralleled developments in our understanding of the human genome and our ability to study genomics at scale; these advancements in genetics have both accompanied and led to those in diabetes treatment. This review will explore the timeline and history of gene discovery and how this has coincided with progress in the fields of genomics. Examples of genetic causes of monogenic diabetes are presented and the continuing expansion of allelic series in these genes and the challenges these now cause for diagnostic interpretation along with opportunities for patient stratification are discussed.

    View details for DOI 10.1530/JOE-21-0067

    View details for PubMedID 34196608

  • There is more than one way to reach type 2 diabetes. Nature metabolism Gloyn, A. L., Powers, A. C. 2021

    View details for DOI 10.1038/s42255-021-00415-6

    View details for PubMedID 34183851

  • The trans-ancestral genomic architecture of glycemic traits. Nature genetics Chen, J., Spracklen, C. N., Marenne, G., Varshney, A., Corbin, L. J., Luan, J., Willems, S. M., Wu, Y., Zhang, X., Horikoshi, M., Boutin, T. S., Magi, R., Waage, J., Li-Gao, R., Chan, K. H., Yao, J., Anasanti, M. D., Chu, A. Y., Claringbould, A., Heikkinen, J., Hong, J., Hottenga, J., Huo, S., Kaakinen, M. A., Louie, T., Marz, W., Moreno-Macias, H., Ndungu, A., Nelson, S. C., Nolte, I. M., North, K. E., Raulerson, C. K., Ray, D., Rohde, R., Rybin, D., Schurmann, C., Sim, X., Southam, L., Stewart, I. D., Wang, C. A., Wang, Y., Wu, P., Zhang, W., Ahluwalia, T. S., Appel, E. V., Bielak, L. F., Brody, J. A., Burtt, N. P., Cabrera, C. P., Cade, B. E., Chai, J. F., Chai, X., Chang, L., Chen, C., Chen, B. H., Chitrala, K. N., Chiu, Y., de Haan, H. G., Delgado, G. E., Demirkan, A., Duan, Q., Engmann, J., Fatumo, S. A., Gayan, J., Giulianini, F., Gong, J. H., Gustafsson, S., Hai, Y., Hartwig, F. P., He, J., Heianza, Y., Huang, T., Huerta-Chagoya, A., Hwang, M. Y., Jensen, R. A., Kawaguchi, T., Kentistou, K. A., Kim, Y. J., Kleber, M. E., Kooner, I. K., Lai, S., Lange, L. A., Langefeld, C. D., Lauzon, M., Li, M., Ligthart, S., Liu, J., Loh, M., Long, J., Lyssenko, V., Mangino, M., Marzi, C., Montasser, M. E., Nag, A., Nakatochi, M., Noce, D., Noordam, R., Pistis, G., Preuss, M., Raffield, L., Rasmussen-Torvik, L. J., Rich, S. S., Robertson, N. R., Rueedi, R., Ryan, K., Sanna, S., Saxena, R., Schraut, K. E., Sennblad, B., Setoh, K., Smith, A. V., Sparso, T., Strawbridge, R. J., Takeuchi, F., Tan, J., Trompet, S., van den Akker, E., van der Most, P. J., Verweij, N., Vogel, M., Wang, H., Wang, C., Wang, N., Warren, H. R., Wen, W., Wilsgaard, T., Wong, A., Wood, A. R., Xie, T., Zafarmand, M. H., Zhao, J., Zhao, W., Amin, N., Arzumanyan, Z., Astrup, A., Bakker, S. J., Baldassarre, D., Beekman, M., Bergman, R. N., Bertoni, A., Bluher, M., Bonnycastle, L. L., Bornstein, S. R., Bowden, D. W., Cai, Q., Campbell, A., Campbell, H., Chang, Y. C., de Geus, E. J., Dehghan, A., Du, S., Eiriksdottir, G., Farmaki, A. E., Franberg, M., Fuchsberger, C., Gao, Y., Gjesing, A. P., Goel, A., Han, S., Hartman, C. A., Herder, C., Hicks, A. A., Hsieh, C., Hsueh, W. A., Ichihara, S., Igase, M., Ikram, M. A., Johnson, W. C., Jorgensen, M. E., Joshi, P. K., Kalyani, R. R., Kandeel, F. R., Katsuya, T., Khor, C. C., Kiess, W., Kolcic, I., Kuulasmaa, T., Kuusisto, J., Lall, K., Lam, K., Lawlor, D. A., Lee, N. R., Lemaitre, R. N., Li, H., Lifelines Cohort Study, Lin, S., Lindstrom, J., Linneberg, A., Liu, J., Lorenzo, C., Matsubara, T., Matsuda, F., Mingrone, G., Mooijaart, S., Moon, S., Nabika, T., Nadkarni, G. N., Nadler, J. L., Nelis, M., Neville, M. J., Norris, J. M., Ohyagi, Y., Peters, A., Peyser, P. A., Polasek, O., Qi, Q., Raven, D., Reilly, D. F., Reiner, A., Rivideneira, F., Roll, K., Rudan, I., Sabanayagam, C., Sandow, K., Sattar, N., Schurmann, A., Shi, J., Stringham, H. M., Taylor, K. D., Teslovich, T. M., Thuesen, B., Timmers, P. R., Tremoli, E., Tsai, M. Y., Uitterlinden, A., van Dam, R. M., van Heemst, D., van Hylckama Vlieg, A., van Vliet-Ostaptchouk, J. V., Vangipurapu, J., Vestergaard, H., Wang, T., Willems van Dijk, K., Zemunik, T., Abecasis, G. R., Adair, L. S., Aguilar-Salinas, C. A., Alarcon-Riquelme, M. E., An, P., Aviles-Santa, L., Becker, D. M., Beilin, L. J., Bergmann, S., Bisgaard, H., Black, C., Boehnke, M., Boerwinkle, E., Bohm, B. O., Bonnelykke, K., Boomsma, D. I., Bottinger, E. P., Buchanan, T. A., Canouil, M., Caulfield, M. J., Chambers, J. C., Chasman, D. I., Chen, Y. I., Cheng, C., Collins, F. S., Correa, A., Cucca, F., de Silva, H. J., Dedoussis, G., Elmstahl, S., Evans, M. K., Ferrannini, E., Ferrucci, L., Florez, J. C., Franks, P. W., Frayling, T. M., Froguel, P., Gigante, B., Goodarzi, M. O., Gordon-Larsen, P., Grallert, H., Grarup, N., Grimsgaard, S., Groop, L., Gudnason, V., Guo, X., Hamsten, A., Hansen, T., Hayward, C., Heckbert, S. R., Horta, B. L., Huang, W., Ingelsson, E., James, P. S., Jarvelin, M., Jonas, J. B., Jukema, J. W., Kaleebu, P., Kaplan, R., Kardia, S. L., Kato, N., Keinanen-Kiukaanniemi, S. M., Kim, B., Kivimaki, M., Koistinen, H. A., Kooner, J. S., Korner, A., Kovacs, P., Kuh, D., Kumari, M., Kutalik, Z., Laakso, M., Lakka, T. A., Launer, L. J., Leander, K., Li, H., Lin, X., Lind, L., Lindgren, C., Liu, S., Loos, R. J., Magnusson, P. K., Mahajan, A., Metspalu, A., Mook-Kanamori, D. O., Mori, T. A., Munroe, P. B., Njolstad, I., O'Connell, J. R., Oldehinkel, A. J., Ong, K. K., Padmanabhan, S., Palmer, C. N., Palmer, N. D., Pedersen, O., Pennell, C. E., Porteous, D. J., Pramstaller, P. P., Province, M. A., Psaty, B. M., Qi, L., Raffel, L. J., Rauramaa, R., Redline, S., Ridker, P. M., Rosendaal, F. R., Saaristo, T. E., Sandhu, M., Saramies, J., Schneiderman, N., Schwarz, P., Scott, L. J., Selvin, E., Sever, P., Shu, X., Slagboom, P. E., Small, K. S., Smith, B. H., Snieder, H., Sofer, T., Sorensen, T. I., Spector, T. D., Stanton, A., Steves, C. J., Stumvoll, M., Sun, L., Tabara, Y., Tai, E. S., Timpson, N. J., Tonjes, A., Tuomilehto, J., Tusie, T., Uusitupa, M., van der Harst, P., van Duijn, C., Vitart, V., Vollenweider, P., Vrijkotte, T. G., Wagenknecht, L. E., Walker, M., Wang, Y. X., Wareham, N. J., Watanabe, R. M., Watkins, H., Wei, W. B., Wickremasinghe, A. R., Willemsen, G., Wilson, J. F., Wong, T., Wu, J., Xiang, A. H., Yanek, L. R., Yengo, L., Yokota, M., Zeggini, E., Zheng, W., Zonderman, A. B., Rotter, J. I., Gloyn, A. L., McCarthy, M. I., Dupuis, J., Meigs, J. B., Scott, R. A., Prokopenko, I., Leong, A., Liu, C., Parker, S. C., Mohlke, K. L., Langenberg, C., Wheeler, E., Morris, A. P., Barroso, I., Meta-Analysis of Glucose and Insulin-related Traits Consortium (MAGIC), de Haan, H. G., van den Akker, E., van der Most, P. J., de Geus, E. J., van Dam, R. M., van Heemst, D., van Hylckama Vlieg, A., van Willems van Dijk, K., de Silva, H. J., van der Harst, P., van Duijn, C. 2021

    Abstract

    Glycemic traits are used to diagnose and monitor type 2 diabetes and cardiometabolic health. To date, most genetic studies of glycemic traits have focused on individuals of European ancestry. Here we aggregated genome-wide association studies comprising up to 281,416 individuals without diabetes (30% non-European ancestry) for whom fasting glucose, 2-h glucose after an oral glucose challenge, glycated hemoglobin and fasting insulin data were available. Trans-ancestry and single-ancestry meta-analyses identified 242loci (99 novel; P<5*10-8), 80% of which had no significant evidence of between-ancestry heterogeneity. Analyses restricted to individuals of European ancestry with equivalent sample size would have led to 24 fewer new loci. Compared with single-ancestry analyses, equivalent-sized trans-ancestry fine-mapping reduced the number of estimated variants in 99% credible sets by a median of 37.5%. Genomic-feature, gene-expression and gene-set analyses revealed distinct biological signatures for each trait, highlighting different underlying biological pathways. Our results increase our understanding of diabetes pathophysiology by using trans-ancestry studies for improved power and resolution.

    View details for DOI 10.1038/s41588-021-00852-9

    View details for PubMedID 34059833

  • Monogenic diabetes: a gateway to precision medicine in diabetes. The Journal of clinical investigation Zhang, H., Colclough, K., Gloyn, A. L., Pollin, T. I. 2021; 131 (3)

    Abstract

    Monogenic diabetes refers to diabetes mellitus (DM) caused by a mutation in a single gene and accounts for approximately 1%-5% of diabetes. Correct diagnosis is clinically critical for certain types of monogenic diabetes, since the appropriate treatment is determined by the etiology of the disease (e.g., oral sulfonylurea treatment of HNF1A/HNF4A-diabetes vs. insulin injections in type 1 diabetes). However, achieving a correct diagnosis requires genetic testing, and the overlapping of the clinical features of monogenic diabetes with those of type 1 and type 2 diabetes has frequently led to misdiagnosis. Improvements in sequencing technology are increasing opportunities to diagnose monogenic diabetes, but challenges remain. In this Review, we describe the types of monogenic diabetes, including common and uncommon types of maturity-onset diabetes of the young, multiple causes of neonatal DM, and syndromic diabetes such as Wolfram syndrome and lipodystrophy. We also review methods of prioritizing patients undergoing genetic testing, and highlight existing challenges facing sequence data interpretation that can be addressed by forming collaborations of expertise and by pooling cases.

    View details for DOI 10.1172/JCI142244

    View details for PubMedID 33529164

  • A Multi-omic Integrative Scheme Characterizes Tissues of Action at Loci Associated with Type 2 Diabetes. American journal of human genetics Torres, J. M., Abdalla, M., Payne, A., Fernandez-Tajes, J., Thurner, M., Nylander, V., Gloyn, A. L., Mahajan, A., McCarthy, M. I. 2020

    Abstract

    Resolving the molecular processes that mediate genetic risk remains a challenge because most disease-associated variants are non-coding and functional characterization of these signals requires knowledge of the specific tissues and cell-types in which they operate. To address this challenge, we developed a framework for integrating tissue-specific gene expression and epigenomic maps to obtain "tissue-of-action" (TOA) scores for each association signal by systematically partitioning posterior probabilities from Bayesian fine-mapping. We applied this scheme to credible set variants for 380 association signals from a recent GWAS meta-analysis of type 2 diabetes (T2D) in Europeans. The resulting tissue profiles underscored a predominant role for pancreatic islets and, to a lesser extent, adipose and liver, particularly among signals with greater fine-mapping resolution. We incorporated resulting TOA scores into a rule-based classifier and validated the tissue assignments through comparison with data from cis-eQTL enrichment, functional fine-mapping, RNA co-expression, and patterns of physiological association. In addition to implicating signals with a single TOA, we found evidence for signals with shared effects in multiple tissues as well as distinct tissue profiles between independent signals within heterogeneous loci. Lastly, we demonstrated that TOA scores can be directly coupled with eQTL colocalization to further resolve effector transcripts at T2D signals. This framework guides mechanistic inference by directing functional validation studies to the most relevant tissues and can gain power as fine-mapping resolution and cell-specific annotations become richer. This method is generalizable to all complex traits with relevant annotation data and is made available as an R package.

    View details for DOI 10.1016/j.ajhg.2020.10.009

    View details for PubMedID 33186544

  • Response to Comment on Misra et al. Homozygous Hypomorphic HNF1A Alleles Are a Novel Cause of Young-Onset Diabetes and Result in Sulfonylurea-Sensitive Diabetes. Diabetes Care 2020;43:909-912. Diabetes care Misra, S., Hassanali, N., Bennett, A. J., Juszczak, A., Caswell, R., Colclough, K., Valabhji, J., Ellard, S., Oliver, N. S., Gloyn, A. L. 2020; 43 (10): e155–e156

    View details for DOI 10.2337/dci20-0033

    View details for PubMedID 32958621

  • Identification of type 2 diabetes loci in 433,540 East Asian individuals. Nature Spracklen, C. N., Horikoshi, M., Kim, Y. J., Lin, K., Bragg, F., Moon, S., Suzuki, K., Tam, C. H., Tabara, Y., Kwak, S., Takeuchi, F., Long, J., Lim, V. J., Chai, J., Chen, C., Nakatochi, M., Yao, J., Choi, H. S., Iyengar, A. K., Perrin, H. J., Brotman, S. M., van de Bunt, M., Gloyn, A. L., Below, J. E., Boehnke, M., Bowden, D. W., Chambers, J. C., Mahajan, A., McCarthy, M. I., Ng, M. C., Petty, L. E., Zhang, W., Morris, A. P., Adair, L. S., Akiyama, M., Bian, Z., Chan, J. C., Chang, L., Chee, M., Chen, Y. I., Chen, Y., Chen, Z., Chuang, L., Du, S., Gordon-Larsen, P., Gross, M., Guo, X., Guo, Y., Han, S., Howard, A., Huang, W., Hung, Y., Hwang, M. Y., Hwu, C., Ichihara, S., Isono, M., Jang, H., Jiang, G., Jonas, J. B., Kamatani, Y., Katsuya, T., Kawaguchi, T., Khor, C., Kohara, K., Lee, M., Lee, N. R., Li, L., Liu, J., Luk, A. O., Lv, J., Okada, Y., Pereira, M. A., Sabanayagam, C., Shi, J., Shin, D. M., So, W. Y., Takahashi, A., Tomlinson, B., Tsai, F., van Dam, R. M., Xiang, Y., Yamamoto, K., Yamauchi, T., Yoon, K., Yu, C., Yuan, J., Zhang, L., Zheng, W., Igase, M., Cho, Y. S., Rotter, J. I., Wang, Y., Sheu, W. H., Yokota, M., Wu, J., Cheng, C., Wong, T., Shu, X., Kato, N., Park, K., Tai, E., Matsuda, F., Koh, W., Ma, R. C., Maeda, S., Millwood, I. Y., Lee, J., Kadowaki, T., Walters, R. G., Kim, B., Mohlke, K. L., Sim, X. 2020

    Abstract

    Meta-analyses of genome-wide association studies (GWAS) have identified more than 240 loci that are associated with type 2 diabetes (T2D)1,2; however, most of these loci have been identified in analyses of individuals with European ancestry. Here, to examine T2D risk in East Asian individuals, we carried out a meta-analysis of GWAS data from 77,418 individuals with T2D and 356,122 healthy control individuals. In the main analysis, we identified 301 distinct association signals at 183 loci, and across T2D association models with and without consideration of body mass index and sex, we identified 61 loci that are newly implicated in predisposition to T2D. Common variants associated with T2D in both East Asian and European populations exhibited strongly correlated effect sizes. Previously undescribed associations include signals in or near GDAP1, PTF1A, SIX3, ALDH2, a microRNA cluster, and genes that affect the differentiation of muscle and adipose cells3. At another locus, expression quantitative trait loci at two overlapping T2D signals affect two genes-NKX6-3 and ANK1-in different tissues4-6. Association studies in diverse populations identify additional loci and elucidate disease-associated genes, biology, and pathways.

    View details for DOI 10.1038/s41586-020-2263-3

    View details for PubMedID 32499647

  • Endocrine-Exocrine Signaling Drives Obesity-Associated Pancreatic Ductal Adenocarcinoma. Cell Chung, K. M., Singh, J., Lawres, L., Dorans, K. J., Garcia, C., Burkhardt, D. B., Robbins, R., Bhutkar, A., Cardone, R., Zhao, X., Babic, A., Vayrynen, S. A., Dias Costa, A., Nowak, J. A., Chang, D. T., Dunne, R. F., Hezel, A. F., Koong, A. C., Wilhelm, J. J., Bellin, M. D., Nylander, V., Gloyn, A. L., McCarthy, M. I., Kibbey, R. G., Krishnaswamy, S., Wolpin, B. M., Jacks, T., Fuchs, C. S., Muzumdar, M. D. 2020

    Abstract

    Obesity is a major modifiable risk factor for pancreatic ductal adenocarcinoma (PDAC), yet how and when obesity contributes to PDAC progression is not well understood. Leveraging an autochthonous mouse model, we demonstrate a causal and reversible role for obesity in early PDAC progression, showing that obesity markedly enhances tumorigenesis, while genetic or dietary induction of weight loss intercepts cancer development. Molecular analyses of human and murine samples define microenvironmental consequences of obesity that foster tumorigenesis rather than new driver gene mutations, including significant pancreatic islet cell adaptation in obesity-associated tumors. Specifically, we identify aberrant beta cell expression of the peptide hormone cholecystokinin (Cck) in response to obesity and show that islet Cck promotes oncogenic Kras-driven pancreatic ductal tumorigenesis. Our studies argue that PDAC progression is driven by local obesity-associated changes in the tumor microenvironment and implicate endocrine-exocrine signaling beyond insulin in PDAC development.

    View details for DOI 10.1016/j.cell.2020.03.062

    View details for PubMedID 32304665

  • From Genetic Association to Molecular Mechanisms for Islet-cell Dysfunction in Type 2 Diabetes JOURNAL OF MOLECULAR BIOLOGY Mattis, K. K., Gloyn, A. L. 2020; 432 (5): 1551–78

    Abstract

    Genome-wide association studies (GWAS) have identified over 400 signals robustly associated with risk for type 2 diabetes (T2D). At the vast majority of these loci, the lead single nucleotide polymorphisms (SNPs) reside in noncoding regions of the genome, which hampers biological inference and translation of genetic discoveries into disease mechanisms. The study of these T2D risk variants in normoglycemic individuals has revealed that a significant proportion are exerting their disease risk through islet-cell dysfunction. The central role of the islet is also demonstrated by numerous studies, which have shown an enrichment of these signals in islet-specific epigenomic annotations. In recent years the emergence of authentic human beta-cell lines, and advances in genome-editing technologies coupled with improved protocols differentiating human pluripotent stem cells into beta-like cells has opened up new opportunities for T2D disease modeling. Here we review the current understanding on the genetic basis of T2D focusing on approaches, which have facilitated the identification of causal variants and their effector transcripts in human islets. We will present examples of functional studies based on animal and conventional cellular systems and highlight the potential of novel stem cell-based T2D disease models.

    View details for DOI 10.1016/j.jmb.2019.12.045

    View details for Web of Science ID 000527955900015

    View details for PubMedID 31945378

  • Editorial Overview: "Islet Biology in Type 2 Diabetes" JOURNAL OF MOLECULAR BIOLOGY Gaisano, H. Y., Jonas, J., Gloyn, A. L. 2020; 432 (5): 1307–9

    View details for DOI 10.1016/j.jmb.2019.12.032

    View details for Web of Science ID 000527955900001

    View details for PubMedID 32005524

  • Insights into pancreatic islet cell dysfunction from type 2 diabetes mellitus genetics. Nature reviews. Endocrinology Krentz, N. A., Gloyn, A. L. 2020

    Abstract

    Type 2 diabetes mellitus (T2DM) is an increasingly prevalent multifactorial disease that has both genetic and environmental risk factors, resulting in impaired glucose homeostasis. Genome-wide association studies (GWAS) have identified over 400 genetic signals that are associated with altered risk of T2DM. Human physiology and epigenomic data support a central role for the pancreatic islet in the pathogenesis of T2DM. This Review focuses on the promises and challenges of moving from genetic associations to molecular mechanisms and highlights efforts to identify the causal variant and effector transcripts at T2DM GWAS susceptibility loci. In addition, we examine current human models that are used to study both beta-cell development and function, including EndoC-beta cell lines and human induced pluripotent stem cell-derived beta-like cells. We use examples of four T2DM susceptibility loci (CDKAL1, MTNR1B, SLC30A8 and PAM) to emphasize how a holistic approach involving genetics, physiology, and cellular and developmental biology can disentangle disease mechanisms at T2DM GWAS signals.

    View details for DOI 10.1038/s41574-020-0325-0

    View details for PubMedID 32099086

  • Deep learning models predict regulatory variants in pancreatic islets and refine type 2 diabetes association signals. eLife Wesolowska-Andersen, A. n., Zhuo Yu, G. n., Nylander, V. n., Abaitua, F. n., Thurner, M. n., Torres, J. M., Mahajan, A. n., Gloyn, A. L., McCarthy, M. I. 2020; 9

    Abstract

    Genome-wide association analyses have uncovered multiple genomic regions associated with T2D, but identification of the causal variants at these remains a challenge. There is growing interest in the potential of deep learning models - which predict epigenome features from DNA sequence - to support inference concerning the regulatory effects of disease-associated variants. Here, we evaluate the advantages of training convolutional neural network (CNN) models on a broad set of epigenomic features collected in a single disease-relevant tissue - pancreatic islets in the case of type 2 diabetes (T2D) - as opposed to models trained on multiple human tissues. We report convergence of CNN-based metrics of regulatory function with conventional approaches to variant prioritization - genetic fine-mapping and regulatory annotation enrichment. We demonstrate that CNN-based analyses can refine association signals at T2D-associated loci and provide experimental validation for one such signal. We anticipate that these approaches will become routine in downstream analyses of GWAS.

    View details for DOI 10.7554/eLife.51503

    View details for PubMedID 31985400

  • Unsupervised Clustering of Missense Variants in HNF1A Using Multidimensional Functional Data Aids Clinical Interpretation. American journal of human genetics Althari, S. n., Najmi, L. A., Bennett, A. J., Aukrust, I. n., Rundle, J. K., Colclough, K. n., Molnes, J. n., Kaci, A. n., Nawaz, S. n., van der Lugt, T. n., Hassanali, N. n., Mahajan, A. n., Molven, A. n., Ellard, S. n., McCarthy, M. I., Bjørkhaug, L. n., Njølstad, P. R., Gloyn, A. L. 2020

    Abstract

    Exome sequencing in diabetes presents a diagnostic challenge because depending on frequency, functional impact, and genomic and environmental contexts, HNF1A variants can cause maturity-onset diabetes of the young (MODY), increase type 2 diabetes risk, or be benign. A correct diagnosis matters as it informs on treatment, progression, and family risk. We describe a multi-dimensional functional dataset of 73 HNF1A missense variants identified in exomes of 12,940 individuals. Our aim was to develop an analytical framework for stratifying variants along the HNF1A phenotypic continuum to facilitate diagnostic interpretation. HNF1A variant function was determined by four different molecular assays. Structure of the multi-dimensional dataset was explored using principal component analysis, k-means, and hierarchical clustering. Weights for tissue-specific isoform expression and functional domain were integrated. Functionally annotated variant subgroups were used to re-evaluate genetic diagnoses in national MODY diagnostic registries. HNF1A variants demonstrated a range of behaviors across the assays. The structure of the multi-parametric data was shaped primarily by transactivation. Using unsupervised learning methods, we obtained high-resolution functional clusters of the variants that separated known causal MODY variants from benign and type 2 diabetes risk variants and led to reclassification of 4% and 9% of HNF1A variants identified in the UK and Norway MODY diagnostic registries, respectively. Our proof-of-principle analyses facilitated informative stratification of HNF1A variants along the continuum, allowing improved evaluation of clinical significance, management, and precision medicine in diabetes clinics. Transcriptional activity appears a superior readout supporting pursuit of transactivation-centric experimental designs for high-throughput functional screens.

    View details for DOI 10.1016/j.ajhg.2020.08.016

    View details for PubMedID 32910913

  • Genetic variant effects on gene expression in human pancreatic islets and their implications for T2D. Nature communications Viñuela, A. n., Varshney, A. n., van de Bunt, M. n., Prasad, R. B., Asplund, O. n., Bennett, A. n., Boehnke, M. n., Brown, A. A., Erdos, M. R., Fadista, J. n., Hansson, O. n., Hatem, G. n., Howald, C. n., Iyengar, A. K., Johnson, P. n., Krus, U. n., MacDonald, P. E., Mahajan, A. n., Manning Fox, J. E., Narisu, N. n., Nylander, V. n., Orchard, P. n., Oskolkov, N. n., Panousis, N. I., Payne, A. n., Stitzel, M. L., Vadlamudi, S. n., Welch, R. n., Collins, F. S., Mohlke, K. L., Gloyn, A. L., Scott, L. J., Dermitzakis, E. T., Groop, L. n., Parker, S. C., McCarthy, M. I. 2020; 11 (1): 4912

    Abstract

    Most signals detected by genome-wide association studies map to non-coding sequence and their tissue-specific effects influence transcriptional regulation. However, key tissues and cell-types required for functional inference are absent from large-scale resources. Here we explore the relationship between genetic variants influencing predisposition to type 2 diabetes (T2D) and related glycemic traits, and human pancreatic islet transcription using data from 420 donors. We find: (a) 7741 cis-eQTLs in islets with a replication rate across 44 GTEx tissues between 40% and 73%; (b) marked overlap between islet cis-eQTL signals and active regulatory sequences in islets, with reduced eQTL effect size observed in the stretch enhancers most strongly implicated in GWAS signal location; (c) enrichment of islet cis-eQTL signals with T2D risk variants identified in genome-wide association studies; and (d) colocalization between 47 islet cis-eQTLs and variants influencing T2D or glycemic traits, including DGKB and TCF7L2. Our findings illustrate the advantages of performing functional and regulatory studies in disease relevant tissues.

    View details for DOI 10.1038/s41467-020-18581-8

    View details for PubMedID 32999275

  • Homozygous Hypomorphic HNF1A Alleles Are a Novel Cause of Young-Onset Diabetes and Result in Sulphonylurea-Sensitive Diabetes. Diabetes care Misra, S. n., Hassanali, N. n., Bennett, A. J., Juszczak, A. n., Caswell, R. n., Colclough, K. n., Valabhji, J. n., Ellard, S. n., Oliver, N. S., Gloyn, A. L. 2020

    Abstract

    Heterozygous loss-of-function mutations in HNF1A cause maturity-onset diabetes of the young (MODY). Affected individuals can be treated with low-dose sulphonylureas. Individuals with homozygous HNF1A mutations causing MODY have not been reported.We phenotyped a kindred with young-onset diabetes and performed molecular genetic testing, a mixed meal tolerance test, a sulphonylurea challenge, and in vitro assays to assess variant protein function.A homozygous HNF1A variant (p.A251T) was identified in three insulin-treated family members diagnosed with diabetes before 20 years of age. Those with the homozygous variant had low hs-CRP levels (0.2-0.8 mg/L), and those tested demonstrated sensitivity to sulphonylurea given at a low dose, completely transitioning off insulin. In silico modeling predicted a variant of unknown significance; however, in vitro studies supported a modest reduction in transactivation potential (79% of that for the wild type; P < 0.05) in the absence of endogenous HNF1A.Homozygous hypomorphic HNF1A variants are a cause of HNF1A-MODY. We thus expand the allelic spectrum of variants in dominant genes causing diabetes.

    View details for DOI 10.2337/dc19-1843

    View details for PubMedID 32001615

  • Analysis of Differentiation Protocols Defines a Common Pancreatic Progenitor Molecular Signature and Guides Refinement of Endocrine Differentiation. Stem cell reports Wesolowska-Andersen, A. n., Jensen, R. R., Alcántara, M. P., Beer, N. L., Duff, C. n., Nylander, V. n., Gosden, M. n., Witty, L. n., Bowden, R. n., McCarthy, M. I., Hansson, M. n., Gloyn, A. L., Honore, C. n. 2020; 14 (1): 138–53

    Abstract

    Several distinct differentiation protocols for deriving pancreatic progenitors (PPs) from human pluripotent stem cells have been described, but it remains to be shown how similar the PPs are across protocols and how well they resemble their in vivo counterparts. Here, we evaluated three differentiation protocols, performed RNA and assay for transposase-accessible chromatin using sequencing on isolated PPs derived with these, and compared them with fetal human pancreas populations. This enabled us to define a shared transcriptional and epigenomic signature of the PPs, including several genes not previously implicated in pancreas development. Furthermore, we identified a significant and previously unappreciated cross-protocol variation of the PPs through multi-omics analysis and demonstrate how such information can be applied to refine differentiation protocols for derivation of insulin-producing beta-like cells. Together, our study highlights the importance of a detailed characterization of defined cell populations derived from distinct differentiation protocols and provides a valuable resource for exploring human pancreatic development.

    View details for DOI 10.1016/j.stemcr.2019.11.010

    View details for PubMedID 31883919

    View details for PubMedCentralID PMC6962645

  • Exocrine or endocrine? A circulating pancreatic elastase that regulates glucose homeostasis. Nature metabolism Gloyn, A. L. 2019; 1 (9): 853-855

    View details for DOI 10.1038/s42255-019-0107-y

    View details for PubMedID 32694741

  • METABOLIC SYNDROME Exocrine or endocrine? A circulating pancreatic elastase that regulates glucose homeostasis NATURE METABOLISM Gloyn, A. L. 2019; 1 (9): 853–55
  • Fostering improved human islet research: a European perspective DIABETOLOGIA Marchetti, P., Schulte, A. M., Marselli, L., Schoniger, E., Bugliani, M., Kramer, W., Overbergh, L., Ullrich, S., Gloyn, A. L., Ibberson, M., Rutter, G., Froguel, P., Groop, L., McCarthy, M. I., Dotta, F., Scharfmann, R., Magnan, C., Eizirik, D. L., Mathieu, C., Cnop, M., Thorens, B., Solimena, M. 2019; 62 (8): 1514–16

    View details for DOI 10.1007/s00125-019-4911-4

    View details for Web of Science ID 000475698900020

    View details for PubMedID 31197398

    View details for PubMedCentralID PMC6647243

  • Developing a network view of type 2 diabetes risk pathways through integration of genetic, genomic and functional data GENOME MEDICINE Fernandez-Tajes, J., Gaulton, K. J., van de Bunt, M., Torres, J., Thurner, M., Mahajan, A., Gloyn, A. L., Lage, K., McCarthy, M. I. 2019; 11: 19

    Abstract

    Genome-wide association studies (GWAS) have identified several hundred susceptibility loci for type 2 diabetes (T2D). One critical, but unresolved, issue concerns the extent to which the mechanisms through which these diverse signals influencing T2D predisposition converge on a limited set of biological processes. However, the causal variants identified by GWAS mostly fall into a non-coding sequence, complicating the task of defining the effector transcripts through which they operate.Here, we describe implementation of an analytical pipeline to address this question. First, we integrate multiple sources of genetic, genomic and biological data to assign positional candidacy scores to the genes that map to T2D GWAS signals. Second, we introduce genes with high scores as seeds within a network optimization algorithm (the asymmetric prize-collecting Steiner tree approach) which uses external, experimentally confirmed protein-protein interaction (PPI) data to generate high-confidence sub-networks. Third, we use GWAS data to test the T2D association enrichment of the "non-seed" proteins introduced into the network, as a measure of the overall functional connectivity of the network.We find (a) non-seed proteins in the T2D protein-interaction network so generated (comprising 705 nodes) are enriched for association to T2D (p = 0.0014) but not control traits, (b) stronger T2D-enrichment for islets than other tissues when we use RNA expression data to generate tissue-specific PPI networks and (c) enhanced enrichment (p = 3.9 × 10- 5) when we combine the analysis of the islet-specific PPI network with a focus on the subset of T2D GWAS loci which act through defective insulin secretion.These analyses reveal a pattern of non-random functional connectivity between candidate causal genes at T2D GWAS loci and highlight the products of genes including YWHAG, SMAD4 or CDK2 as potential contributors to T2D-relevant islet dysfunction. The approach we describe can be applied to other complex genetic and genomic datasets, facilitating integration of diverse data types into disease-associated networks.

    View details for DOI 10.1186/s13073-019-0628-8

    View details for Web of Science ID 000462609800001

    View details for PubMedID 30914061

    View details for PubMedCentralID PMC6436236

  • Loss of ZnT8 function protects against diabetes by enhanced insulin secretion. Nature genetics Dwivedi, O. P., Lehtovirta, M. n., Hastoy, B. n., Chandra, V. n., Krentz, N. A., Kleiner, S. n., Jain, D. n., Richard, A. M., Abaitua, F. n., Beer, N. L., Grotz, A. n., Prasad, R. B., Hansson, O. n., Ahlqvist, E. n., Krus, U. n., Artner, I. n., Suoranta, A. n., Gomez, D. n., Baras, A. n., Champon, B. n., Payne, A. J., Moralli, D. n., Thomsen, S. K., Kramer, P. n., Spiliotis, I. n., Ramracheya, R. n., Chabosseau, P. n., Theodoulou, A. n., Cheung, R. n., van de Bunt, M. n., Flannick, J. n., Trombetta, M. n., Bonora, E. n., Wolheim, C. B., Sarelin, L. n., Bonadonna, R. C., Rorsman, P. n., Davies, B. n., Brosnan, J. n., McCarthy, M. I., Otonkoski, T. n., Lagerstedt, J. O., Rutter, G. A., Gromada, J. n., Gloyn, A. L., Tuomi, T. n., Groop, L. n. 2019; 51 (11): 1596–1606

    Abstract

    A rare loss-of-function allele p.Arg138* in SLC30A8 encoding the zinc transporter 8 (ZnT8), which is enriched in Western Finland, protects against type 2 diabetes (T2D). We recruited relatives of the identified carriers and showed that protection was associated with better insulin secretion due to enhanced glucose responsiveness and proinsulin conversion, particularly when compared with individuals matched for the genotype of a common T2D-risk allele in SLC30A8, p.Arg325. In genome-edited human induced pluripotent stem cell (iPSC)-derived β-like cells, we establish that the p.Arg138* allele results in reduced SLC30A8 expression due to haploinsufficiency. In human β cells, loss of SLC30A8 leads to increased glucose responsiveness and reduced KATP channel function similar to isolated islets from carriers of the T2D-protective allele p.Trp325. These data position ZnT8 as an appealing target for treatment aimed at maintaining insulin secretion capacity in T2D.

    View details for DOI 10.1038/s41588-019-0513-9

    View details for PubMedID 31676859

    View details for PubMedCentralID PMC6858874

  • Plasma Fucosylated Glycans and C-Reactive Protein as Biomarkers of HNF1A-MODY in Young Adult-Onset Nonautoimmune Diabetes DIABETES CARE Juszczak, A., Pavic, T., Vuckovic, F., Bennett, A. J., Shah, N., Medvidovic, E., Groves, C. J., Sekerija, M., Chandler, K., Burrows, C., Putarek, N., Lovrencic, M., Knezevic, J., James, T. J., Gloyn, A. L., Lauc, G., McCarthy, M. I., Owen, K. R., Gornik, O. 2019; 42 (1): 17–26

    Abstract

    Maturity-onset diabetes of the young (MODY) due to variants in HNF1A is the most common type of monogenic diabetes. Frequent misdiagnosis results in missed opportunity to use sulfonylureas as first-line treatment. A nongenetic biomarker could improve selection of subjects for genetic testing and increase diagnosis rates. We previously reported that plasma levels of antennary fucosylated N-glycans and high-sensitivity C-reactive protein (hs-CRP) are reduced in individuals with HNF1A-MODY. In this study, we examined the potential use of N-glycans and hs-CRP in discriminating individuals with damaging HNF1A alleles from those without HNF1A variants in an unselected population of young adults with nonautoimmune diabetes.We analyzed the plasma N-glycan profile, measured hs-CRP, and sequenced HNF1A in 989 individuals with diabetes diagnosed when younger than age 45, persistent endogenous insulin production, and absence of pancreatic autoimmunity. Systematic assessment of rare HNF1A variants was performed.We identified 29 individuals harboring 25 rare HNF1A alleles, of which 3 were novel, and 12 (in 16 probands) were considered pathogenic. Antennary fucosylated N-glycans and hs-CRP were able to differentiate subjects with damaging HNF1A alleles from those without rare HNF1A alleles. Glycan GP30 had a receiver operating characteristic curve area under the curve (AUC) of 0.90 (88% sensitivity, 80% specificity, cutoff 0.70%), whereas hs-CRP had an AUC of 0.83 (88% sensitivity, 69% specificity, cutoff 0.81 mg/L).Half of rare HNF1A sequence variants do not cause MODY. N-glycan profile and hs-CRP could both be used as tools, alone or as adjuncts to existing pathways, for identifying individuals at high risk of carrying a damaging HNF1A allele.

    View details for DOI 10.2337/dc18-0422

    View details for Web of Science ID 000453904900013

    View details for PubMedID 30455330

  • A CRISPR/Cas9 genome editing pipeline in the EndoC-βH1 cell line to study genes implicated in beta cell function. Wellcome open research Grotz, A. K., Abaitua, F. n., Navarro-Guerrero, E. n., Hastoy, B. n., Ebner, D. n., Gloyn, A. L. 2019; 4: 150

    Abstract

    Type 2 diabetes (T2D) is a global pandemic with a strong genetic component, but most causal genes influencing the disease risk remain unknown. It is clear, however, that the pancreatic beta cell is central to T2D pathogenesis. In vitro gene-knockout (KO) models to study T2D risk genes have so far focused on rodent beta cells. However, there are important structural and functional differences between rodent and human beta cell lines. With that in mind, we have developed a robust pipeline to create a stable CRISPR/Cas9 KO in an authentic human beta cell line (EndoC-βH1). The KO pipeline consists of a dual lentiviral sgRNA strategy and we targeted three genes ( INS, IDE, PAM) as a proof of concept. We achieved a significant reduction in mRNA levels and complete protein depletion of all target genes. Using this dual sgRNA strategy, up to 94 kb DNA were cut out of the target genes and the editing efficiency of each sgRNA exceeded >87.5%. Sequencing of off-targets showed no unspecific editing. Most importantly, the pipeline did not affect the glucose-responsive insulin secretion of the cells. Interestingly, comparison of KO cell lines for NEUROD1 and SLC30A8 with siRNA-mediated knockdown (KD) approaches demonstrate phenotypic differences. NEUROD1-KO cells were not viable and displayed elevated markers for ER stress and apoptosis. NEUROD1-KD, however, only had a modest elevation, by 34%, in the pro-apoptotic transcription factor CHOP and a gene expression profile indicative of chronic ER stress without evidence of elevated cell death. On the other hand, SLC30A8-KO cells demonstrated no reduction in K ATP channel gene expression in contrast to siRNA silencing. Overall, this strategy to efficiently create stable KO in the human beta cell line EndoC-βH1 will allow for a better understanding of genes involved in beta cell dysfunction, their underlying functional mechanisms and T2D pathogenesis.

    View details for DOI 10.12688/wellcomeopenres.15447.1

    View details for PubMedID 31976379

    View details for PubMedCentralID PMC6961417

  • Translational genomics and precision medicine: Moving from the lab to the clinic. Science (New York, N.Y.) Zeggini, E. n., Gloyn, A. L., Barton, A. C., Wain, L. V. 2019; 365 (6460): 1409–13

    Abstract

    Translational genomics aims to improve human health by building on discoveries made through genetics research and applying them in the clinical setting. This progress has been made possible by technological advances in genomics and analytics and by the digital revolution. Such advances should enable the development of prognostic markers, tailored interventions, and the design of prophylactic preventive approaches. We are at the cusp of predicting disease risk for some disorders by means of polygenic risk scores integrated with classical epidemiological risk factors. This should lead to better risk stratification and clinical decision-making. A deeper understanding of the link between genome-wide sequence and association with well-characterized phenotypes will empower the development of biomarkers to aid diagnosis, inform disease progression trajectories, and allow better targeting of treatments to those patients most likely to respond.

    View details for DOI 10.1126/science.aax4588

    View details for PubMedID 31604268

  • Fine-mapping type 2 diabetes loci to single-variant resolution using high-density imputation and islet-specific epigenome maps. Nature genetics Mahajan, A., Taliun, D., Thurner, M., Robertson, N. R., Torres, J. M., Rayner, N. W., Payne, A. J., Steinthorsdottir, V., Scott, R. A., Grarup, N., Cook, J. P., Schmidt, E. M., Wuttke, M., Sarnowski, C., Magi, R., Nano, J., Gieger, C., Trompet, S., Lecoeur, C., Preuss, M. H., Prins, B. P., Guo, X., Bielak, L. F., Below, J. E., Bowden, D. W., Chambers, J. C., Kim, Y. J., Ng, M. C., Petty, L. E., Sim, X., Zhang, W., Bennett, A. J., Bork-Jensen, J., Brummett, C. M., Canouil, M., Ec Kardt, K., Fischer, K., Kardia, S. L., Kronenberg, F., Lall, K., Liu, C., Locke, A. E., Luan, J., Ntalla, I., Nylander, V., Schonherr, S., Schurmann, C., Yengo, L., Bottinger, E. P., Brandslund, I., Christensen, C., Dedoussis, G., Florez, J. C., Ford, I., Franco, O. H., Frayling, T. M., Giedraitis, V., Hackinger, S., Hattersley, A. T., Herder, C., Ikram, M. A., Ingelsson, M., Jorgensen, M. E., Jorgensen, T., Kriebel, J., Kuusisto, J., Ligthart, S., Lindgren, C. M., Linneberg, A., Lyssenko, V., Mamakou, V., Meitinger, T., Mohlke, K. L., Morris, A. D., Nadkarni, G., Pankow, J. S., Peters, A., Sattar, N., Stancakova, A., Strauch, K., Taylor, K. D., Thorand, B., Thorleifsson, G., Thorsteinsdottir, U., Tuomilehto, J., Witte, D. R., Dupuis, J., Peyser, P. A., Zeggini, E., Loos, R. J., Froguel, P., Ingelsson, E., Lind, L., Groop, L., Laakso, M., Collins, F. S., Jukema, J. W., Palmer, C. N., Grallert, H., Metspalu, A., Dehghan, A., Kottgen, A., Abecasis, G. R., Meigs, J. B., Rotter, J. I., Marchini, J., Pedersen, O., Hansen, T., Langenberg, C., Wareham, N. J., Stefansson, K., Gloyn, A. L., Morris, A. P., Boehnke, M., McCarthy, M. I. 2018

    Abstract

    We expanded GWAS discovery for type 2 diabetes (T2D) by combining data from 898,130 European-descent individuals (9% cases), after imputation to high-density reference panels. With these data, we (i) extend the inventory of T2D-risk variants (243 loci, 135 newly implicated in T2D predisposition, comprising 403 distinct association signals); (ii) enrich discovery of lower-frequency risk alleles (80 index variants with minor allele frequency <5%, 14 with estimated allelic odds ratio >2); (iii) substantially improve fine-mapping of causal variants (at 51 signals, one variant accounted for >80% posterior probability of association (PPA)); (iv) extend fine-mapping through integration of tissue-specific epigenomic information (islet regulatory annotations extend the number of variants with PPA >80% to 73); (v) highlight validated therapeutic targets (18 genes with associations attributable to coding variants); and (vi) demonstrate enhanced potential for clinical translation (genome-wide chip heritability explains 18% of T2D risk; individuals in the extremes of a T2D polygenic risk score differ more than ninefold in prevalence).

    View details for PubMedID 30297969

  • Understanding human fetal pancreas development using subpopulation sorting, RNA sequencing and single-cell profiling DEVELOPMENT Ramond, C., Beydag-Tasoz, B., Azad, A., van de Bunt, M., Petersen, M., Beer, N. L., Glaser, N., Berthault, C., Gloyn, A. L., Hansson, M., McCarthy, M., Honore, C., Grapin-Botton, A., Scharfmann, R. 2018; 145 (16)

    Abstract

    To decipher the populations of cells present in the human fetal pancreas and their lineage relationships, we developed strategies to isolate pancreatic progenitors, endocrine progenitors and endocrine cells. Transcriptome analysis of the individual populations revealed a large degree of conservation among vertebrates in the drivers of gene expression changes that occur at different steps of differentiation, although notably, sometimes, different members of the same gene family are expressed. The transcriptome analysis establishes a resource to identify novel genes and pathways involved in human pancreas development. Single-cell profiling further captured intermediate stages of differentiation and enabled us to decipher the sequence of transcriptional events occurring during human endocrine differentiation. Furthermore, we evaluate how well individual pancreatic cells derived in vitro from human pluripotent stem cells mirror the natural process occurring in human fetuses. This comparison uncovers a few differences at the progenitor steps, a convergence at the steps of endocrine induction, and the current inability to fully resolve endocrine cell subtypes in vitro.

    View details for DOI 10.1242/dev.165480

    View details for Web of Science ID 000443499500010

    View details for PubMedID 30042179

    View details for PubMedCentralID PMC6124547

  • Patterns of differential gene expression in a cellular model of human islet development, and relationship to type 2 diabetes predisposition DIABETOLOGIA Perez-Alcantara, M., Honore, C., Wesolowska-Andersen, A., Gloyn, A. L., McCarthy, M. I., Hansson, M., Beer, N. L., van de Bunt, M. 2018; 61 (7): 1614–22

    Abstract

    Most type 2 diabetes-associated genetic variants identified via genome-wide association studies (GWASs) appear to act via the pancreatic islet. Observed defects in insulin secretion could result from an impact of these variants on islet development and/or the function of mature islets. Most functional studies have focused on the latter, given limitations regarding access to human fetal islet tissue. Capitalising upon advances in in vitro differentiation, we characterised the transcriptomes of human induced pluripotent stem cell (iPSC) lines differentiated along the pancreatic endocrine lineage, and explored the contribution of altered islet development to the pathogenesis of type 2 diabetes.We performed whole-transcriptome RNA sequencing of human iPSC lines from three independent donors, at baseline and at seven subsequent stages during in vitro islet differentiation. Differentially expressed genes (q < 0.01, log2 fold change [FC] > 1) were assigned to the stages at which they were most markedly upregulated. We used these data to characterise upstream transcription factors directing different stages of development, and to explore the relationship between RNA expression profiles and genes mapping to type 2 diabetes GWAS signals.We identified 9409 differentially expressed genes across all stages, including many known markers of islet development. Integration of differential expression data with information on transcription factor motifs highlighted the potential contribution of REST to islet development. Over 70% of genes mapping within type 2 diabetes-associated credible intervals showed peak differential expression during islet development, and type 2 diabetes GWAS loci of largest effect (including TCF7L2; log2FC = 1.2; q = 8.5 × 10-10) were notably enriched in genes differentially expressed at the posterior foregut stage (q = 0.002), as calculated by gene set enrichment analyses. In a complementary analysis of enrichment, genes differentially expressed in the final, beta-like cell stage of in vitro differentiation were significantly enriched (hypergeometric test, permuted p value <0.05) for genes within the credible intervals of type 2 diabetes GWAS loci.The present study characterises RNA expression profiles during human islet differentiation, identifies potential transcriptional regulators of the differentiation process, and suggests that the inherited predisposition to type 2 diabetes is partly mediated through modulation of islet development.Sequence data for this study has been deposited at the European Genome-phenome Archive (EGA), under accession number EGAS00001002721.

    View details for DOI 10.1007/s00125-018-4612-4

    View details for Web of Science ID 000434250500013

    View details for PubMedID 29675560

    View details for PubMedCentralID PMC6354904

  • NKX6.1 induced pluripotent stem cell reporter lines for isolation and analysis of functionally relevant neuronal and pancreas populations STEM CELL RESEARCH Gupta, S., Wesolowska-Andersen, A., Ringgaard, A. K., Jaiswal, H., Song, L., Hastoy, B., Ingvorsen, C., Taheri-Ghahfarokhi, A., Magnusson, B., Maresca, M., Jensen, R. R., Beer, N. L., Fels, J. J., Grunnet, L. G., Thomas, M. K., Gloyn, A. L., Hicks, R., McCarthy, M. I., Hansson, M., Honore, C. 2018; 29: 220–31

    Abstract

    Recent studies have reported significant advances in the differentiation of human pluripotent stem cells to clinically relevant cell types such as the insulin producing beta-like cells and motor neurons. However, many of the current differentiation protocols lead to heterogeneous cell cultures containing cell types other than the targeted cell fate. Genetically modified human pluripotent stem cells reporting the expression of specific genes are of great value for differentiation protocol optimization and for the purification of relevant cell populations from heterogeneous cell cultures. Here we present the generation of human induced pluripotent stem cell (iPSC) lines with a GFP reporter inserted in the endogenous NKX6.1 locus. Characterization of the reporter lines demonstrated faithful GFP labelling of NKX6.1 expression during pancreas and motor neuron differentiation. Cell sorting and gene expression profiling by RNA sequencing revealed that NKX6.1-positive cells from pancreatic differentiations closely resemble human beta cells. Furthermore, functional characterization of the isolated cells demonstrated that glucose-stimulated insulin secretion is mainly confined to the NKX6.1-positive cells. We expect that the NKX6.1-GFP iPSC lines and the results presented here will contribute to the further refinement of differentiation protocols and characterization of hPSC-derived beta cells and motor neurons for disease modelling and cell replacement therapies.

    View details for DOI 10.1016/j.scr.2018.04.010

    View details for Web of Science ID 000434978900038

    View details for PubMedID 29734117

  • Regulatory variants at KLF14 influence type 2 diabetes risk via a female-specific effect on adipocyte size and body composition NATURE GENETICS Small, K. S., Todorcevic, M., Civelek, M., Moustafa, J., Wang, X., Simon, M. M., Fernandez-Tajes, J., Mahajan, A., Horikoshi, M., Hugill, A., Glastonbury, C. A., Quaye, L., Neville, M. J., Sethi, S., Yon, M., Pan, C., Che, N., Vinuela, A., Tsai, P., Nag, A., Buil, A., Thorleifsson, G., Raghavan, A., Ding, Q., Morris, A. P., Bell, J. T., Thorsteinsdottir, U., Stefansson, K., Laakso, M., Dahlman, I., Arner, P., Gloyn, A. L., Musunuru, K., Lusis, A. J., Cox, R. D., Karpe, F., McCarthy, M. I. 2018; 50 (4): 572-+

    Abstract

    Individual risk of type 2 diabetes (T2D) is modified by perturbations to the mass, distribution and function of adipose tissue. To investigate the mechanisms underlying these associations, we explored the molecular, cellular and whole-body effects of T2D-associated alleles near KLF14. We show that KLF14 diabetes-risk alleles act in adipose tissue to reduce KLF14 expression and modulate, in trans, the expression of 385 genes. We demonstrate, in human cellular studies, that reduced KLF14 expression increases pre-adipocyte proliferation but disrupts lipogenesis, and in mice, that adipose tissue-specific deletion of Klf14 partially recapitulates the human phenotype of insulin resistance, dyslipidemia and T2D. We show that carriers of the KLF14 T2D risk allele shift body fat from gynoid stores to abdominal stores and display a marked increase in adipocyte cell size, and that these effects on fat distribution, and the T2D association, are female specific. The metabolic risk associated with variation at this imprinted locus depends on the sex both of the subject and of the parent from whom the risk allele derives.

    View details for DOI 10.1038/s41588-018-0088-x

    View details for Web of Science ID 000429529300017

    View details for PubMedID 29632379

    View details for PubMedCentralID PMC5935235

  • A Partial Loss-of-Function Variant in AKT2 Is Associated With Reduced Insulin-Mediated Glucose Uptake in Multiple Insulin-Sensitive Tissues: A Genotype-Based Callback Positron Emission Tomography Study DIABETES Latva-Rasku, A., Honka, M., Stancakova, A., Koistinen, H. A., Kuusisto, J., Guan, L., Manning, A. K., Stringham, H., Gloyn, A. L., Lindgren, C. M., Collins, F. S., Mohlke, K. L., Scott, L. J., Karjalainen, T., Nummenmaa, L., Boehnke, M., Nuutila, P., Laakso, M., T2D-GENES Consortium 2018; 67 (2): 334–42

    Abstract

    Rare fully penetrant mutations in AKT2 are an established cause of monogenic disorders of glucose metabolism. Recently, a novel partial loss-of-function AKT2 coding variant (p.Pro50Thr) was identified that is nearly specific to Finns (frequency 1.1%), with the low-frequency allele associated with an increase in fasting plasma insulin level and risk of type 2 diabetes. The effects of the p.Pro50Thr AKT2 variant (p.P50T/AKT2) on insulin-stimulated glucose uptake (GU) in the whole body and in different tissues have not previously been investigated. We identified carriers (N = 20) and matched noncarriers (N = 25) for this allele in the population-based Metabolic Syndrome in Men (METSIM)study and invited these individuals back for positron emission tomography study with [18F]-fluorodeoxyglucose during euglycemic hyperinsulinemia. When we compared p.P50T/AKT2 carriers to noncarriers, we found a 39.4% reduction in whole-body GU (P = 0.006) and a 55.6% increase in the rate of endogenous glucose production (P = 0.038). We found significant reductions in GU in multiple tissues-skeletal muscle (36.4%), liver (16.1%), brown adipose (29.7%), and bone marrow (32.9%)-and increases of 16.8-19.1% in seven tested brain regions. These data demonstrate that the p.P50T substitution of AKT2 influences insulin-mediated GU in multiple insulin-sensitive tissues and may explain, at least in part, the increased risk of type 2 diabetes in p.P50T/AKT2 carriers.

    View details for DOI 10.2337/db17-1142

    View details for Web of Science ID 000426034500016

    View details for PubMedID 29141982

    View details for PubMedCentralID PMC5780065

  • Sequence data and association statistics from 12,940 type 2 diabetes cases and controls (vol 4, 170179, 2017) SCIENTIFIC DATA Flannick, J., Fuchsberger, C., Mahajan, A., Teslovich, T. M., Agarwala, V., Gaulton, K. J., Caulkins, L., Koesterer, R., Ma, C., Moutsianas, L., McCarthy, D. J., Rivas, M. A., Perry, J. B., Sim, X., Blackwell, T. W., Robertson, N. R., Rayner, N., Cingolani, P., Locke, A. E., Tajes, J., Highland, H. M., Dupuis, J., Chines, P. S., Lindgren, C. M., Hartl, C., Jackson, A. U., Chen, H., Huyghe, J. R., van de Bunt, M., Pearson, R. D., Kumar, A., Mueller-Nurasyid, M., Grarup, N., Stringham, H. M., Gamazon, E. R., Lee, J., Chen, Y., Scott, R. A., Below, J. E., Chen, P., Huang, J., Go, M., Stitzel, M. L., Pasko, D., Parker, S. J., Varga, T. V., Green, T., Beer, N. L., Day-Williams, A. G., Ferreira, T., Fingerlin, T., Horikoshi, M., Hu, C., Huh, I., Ikram, M., Kim, B., Kim, Y., Kim, Y., Kwon, M., Lee, J., Lee, S., Lin, K., Maxwell, T. J., Nagai, Y., Wang, X., Welch, R. P., Yoon, J., Zhang, W., Barzilai, N., Voight, B. F., Han, B., Jenkinson, C. P., Kuulasmaa, T., Kuusisto, J., Manning, A., Ng, M. Y., Palmer, N. D., Balkau, B., Stancakova, A., Abboud, H. E., Boeing, H., Giedraitis, V., Prabhakaran, D., Gottesman, O., Scott, J., Carey, J., Kwan, P., Grant, G., Smith, J. D., Neale, B. M., Purcell, S., Butterworth, A. S., Howson, J. M., Lee, H., Lu, Y., Kwak, S., Zhao, W., Danesh, J., Lam, V. L., Park, K., Saleheen, D., So, W., Tam, C. T., Afzal, U., Aguilar, D., Arya, R., Aung, T., Chan, E., Navarro, C., Cheng, C., Palli, D., Correa, A., Curran, J. E., Rybin, D., Farook, V. S., Fowler, S. P., Freedman, B. I., Griswold, M., Hale, D., Hicks, P. J., Khor, C., Kumar, S., Lehne, B., Thuillier, D., Lim, W., Liu, J., Loh, M., Musani, S. K., Puppala, S., Scott, W. R., Yengo, L., Tan, S., Taylor, H. A., Thameem, F., Wilson, G., Wong, T., Njolstad, P., Levy, J. C., Mangino, M., Bonnycastle, L. L., Schwarzmayr, T., Fadista, J., Surdulescu, G. L., Herder, C., Groves, C. J., Wieland, T., Bork-Jensen, J., Brandslund, I., Christensen, C., Koistinen, H. A., Doney, A. F., Kinnunen, L., Esko, T., Farmer, A. J., Hakaste, L., Hodgkiss, D., Kravic, J., Lyssenko, V., Hollensted, M., Jorgensen, M. E., Jorgensen, T., Ladenvall, C., Justesen, J., Karajamaki, A., Kriebel, J., Rathmann, W., Lannfelt, L., Lauritzen, T., Narisu, N., Linneberg, A., Melander, O., Milani, L., Neville, M., Orho-Melander, M., Qi, L., Qi, Q., Roden, M., Rolandsson, O., Swift, A., Rosengren, A. H., Stirrups, K., Wood, A. R., Mihailov, E., Blancher, C., Carneiro, M. O., Maguire, J., Poplin, R., Shakir, K., Fennell, T., DePristo, M., de Angelis, M., Deloukas, P., Gjesing, A. P., Jun, G., Nilsson, P. M., Murphy, J., Onofrio, R., Thorand, B., Hansen, T., Meisinger, C., Hu, F. B., Isomaa, B., Karpe, F., Liang, L., Peters, A., Huth, C., O'Rahilly, S. P., Palmer, C. A., Pedersen, O., Rauramaa, R., Tuomilehto, J., Salomaa, V., Watanabe, R. M., Syvanen, A., Bergman, R. N., Bharadwaj, D., Bottinger, E. P., Cho, Y., Chandak, G. R., Chan, J., Chia, K., Daly, M. J., Ebrahim, S. B., Langenberg, C., Elliott, P., Jablonski, K. A., Lehman, D. M., Jia, W., Ma, R. W., Pollin, T. I., Sandhu, M., Tandon, N., Froguel, P., Barroso, I., Teo, Y., Zeggini, E., Loos, R. F., Small, K. S., Ried, J. S., DeFronzo, R. A., Grallert, H., Glaser, B., Metspalu, A., Wareham, N. J., Walker, M., Banks, E., Gieger, C., Ingelsson, E., Im, H., Illig, T., Franks, P. W., Buck, G., Trakalo, J., Buck, D., Prokopenko, I., Magi, R., Lind, L., Farjoun, Y., Owen, K. R., Gloyn, A. L., Strauch, K., Tuomi, T., Kooner, J., Lee, J., Park, T., Donnelly, P., Morris, A. D., Hattersley, A. T., Bowden, D. W., Collins, F. S., Atzmon, G., Chambers, J. C., Spector, T. D., Laakso, M., Strom, T. M., Bell, G. I., Blangero, J., Duggirala, R., Tai, E. S., McVean, G., Hanis, C. L., Wilson, J. G., Seielstad, M., Frayling, T. M., Meigs, J. B., Cox, N. J., Sladek, R., Lander, E. S., Gabriel, S., Mohlke, K. L., Meitinger, T., Groop, L., Abecasis, G., Scott, L. J., Morris, A. P., Kang, H., Altshuler, D., Burtt, N. P., Florez, J. C., Boehnke, M., McCarthy, M. I. 2018; 5: 180002

    Abstract

    This corrects the article DOI: 10.1038/sdata.2017.179.

    View details for DOI 10.1038/sdata.2018.2

    View details for Web of Science ID 000423058900001

    View details for PubMedID 29360107

    View details for PubMedCentralID PMC5779067

  • Type 2 diabetes risk alleles in PAM impact insulin release from human pancreatic β-cells. Nature genetics Thomsen, S. K., Raimondo, A. n., Hastoy, B. n., Sengupta, S. n., Dai, X. Q., Bautista, A. n., Censin, J. n., Payne, A. J., Umapathysivam, M. M., Spigelman, A. F., Barrett, A. n., Groves, C. J., Beer, N. L., Manning Fox, J. E., McCarthy, M. I., Clark, A. n., Mahajan, A. n., Rorsman, P. n., MacDonald, P. E., Gloyn, A. L. 2018; 50 (8): 1122–31

    Abstract

    The molecular mechanisms underpinning susceptibility loci for type 2 diabetes (T2D) remain poorly understood. Coding variants in peptidylglycine α-amidating monooxygenase (PAM) are associated with both T2D risk and insulinogenic index. Here, we demonstrate that the T2D risk alleles impact negatively on overall PAM activity via defects in expression and catalytic function. PAM deficiency results in reduced insulin content and altered dynamics of insulin secretion in a human β-cell model and primary islets from cadaveric donors. Thus, our results demonstrate a role for PAM in β-cell function, and establish molecular mechanisms for T2D risk alleles at this locus.

    View details for DOI 10.1038/s41588-018-0173-1

    View details for PubMedID 30054598

    View details for PubMedCentralID PMC6237273

  • Maturity onset diabetes of the young due to HNF1A variants in Croatia BIOCHEMIA MEDICA Pavic, T., Juszczak, A., Medvidovic, E., Burrows, C., Sekerija, M., Bennett, A. J., Knezevic, J., Gloyn, A. L., Lauc, G., McCarthy, M., Gornik, O., Owen, K. R. 2018; 28 (2): 020703

    Abstract

    Maturity onset diabetes of the young due to HNF1A mutations (HNF1A-MODY) is the most frequent form of monogenic diabetes in adults. It is often misdiagnosed as type 1 or type 2 diabetes, but establishing genetic diagnosis is important, as treatment differs from the common types of diabetes. HNF1A-MODY has not been investigated in Croatia before due to limited access to genetic testing. In this study we aimed to describe the characteristics of young adults diagnosed with diabetes before the age of 45 years, who have rare HNF1A allele variants, and estimate the prevalence of HNF1A-MODY in Croatia.We recruited 477 C-peptide positive and beta cell antibody negative subjects through the Croatian Diabetes Registry. HNF1A was sequenced for all participants and systematic assessment of the variants found was performed. The prevalence of HNF1A-MODY was calculated in the study group and results extrapolated to estimate the proportion of diabetic individuals with HNF1A-MODY in Croatia and the population prevalence.Our study identified 13 individuals harbouring rare HNF1A allelic variants. After systematic assessment, 8 were assigned a diagnosis of HNF1A-MODY. Two individuals were able to discontinue insulin treatment following the diagnosis. We estimated that HNF1A-MODY in Croatia has a prevalence of 66 (95% CI 61 - 72) cases per million.The estimated prevalence of HNF1A-MODY in Croatia is similar to that reported in other European countries. Finding cases lead to important treatment changes for patients. This strongly supports the introduction of diagnostic genetic testing for monogenic diabetes in Croatia.

    View details for DOI 10.11613/BM.2018.020703

    View details for Web of Science ID 000436129200008

    View details for PubMedID 29666556

    View details for PubMedCentralID PMC5898959

  • Electrophysiological properties of human beta-cell lines EndoC-βH1 and -βH2 conform with human beta-cells. Scientific reports Hastoy, B. n., Godazgar, M. n., Clark, A. n., Nylander, V. n., Spiliotis, I. n., van de Bunt, M. n., Chibalina, M. V., Barrett, A. n., Burrows, C. n., Tarasov, A. I., Scharfmann, R. n., Gloyn, A. L., Rorsman, P. n. 2018; 8 (1): 16994

    Abstract

    Limited access to human islets has prompted the development of human beta cell models. The human beta cell lines EndoC-βH1 and EndoC-βH2 are increasingly used by the research community. However, little is known of their electrophysiological and secretory properties. Here, we monitored parameters that constitute the glucose-triggering pathway of insulin release. Both cell lines respond to glucose (6 and 20 mM) with 2- to 3-fold stimulation of insulin secretion which correlated with an elevation of [Ca2+]i, membrane depolarisation and increased action potential firing. Similar to human primary beta cells, KATP channel activity is low at 1 mM glucose and is further reduced upon increasing glucose concentration; an effect that was mimicked by the KATP channel blocker tolbutamide. The upstroke of the action potentials reflects the activation of Ca2+ channels with some small contribution of TTX-sensitive Na+ channels. The repolarisation involves activation of voltage-gated Kv2.2 channels and large-conductance Ca2+-activated K+ channels. Exocytosis presented a similar kinetics to human primary beta cells. The ultrastructure of these cells shows insulin vesicles composed of an electron-dense core surrounded by a thin clear halo. We conclude that the EndoC-βH1 and -βH2 cells share many features of primary human β-cells and thus represent a useful experimental model.

    View details for DOI 10.1038/s41598-018-34743-7

    View details for PubMedID 30451893

    View details for PubMedCentralID PMC6242937

  • Integration of human pancreatic islet genomic data refines regulatory mechanisms at Type 2 Diabetes susceptibility loci. eLife Thurner, M. n., van de Bunt, M. n., Torres, J. M., Mahajan, A. n., Nylander, V. n., Bennett, A. J., Gaulton, K. J., Barrett, A. n., Burrows, C. n., Bell, C. G., Lowe, R. n., Beck, S. n., Rakyan, V. K., Gloyn, A. L., McCarthy, M. I. 2018; 7

    Abstract

    Human genetic studies have emphasised the dominant contribution of pancreatic islet dysfunction to development of Type 2 Diabetes (T2D). However, limited annotation of the islet epigenome has constrained efforts to define the molecular mechanisms mediating the, largely regulatory, signals revealed by Genome-Wide Association Studies (GWAS). We characterised patterns of chromatin accessibility (ATAC-seq, n = 17) and DNA methylation (whole-genome bisulphite sequencing, n = 10) in human islets, generating high-resolution chromatin state maps through integration with established ChIP-seq marks. We found enrichment of GWAS signals for T2D and fasting glucose was concentrated in subsets of islet enhancers characterised by open chromatin and hypomethylation, with the former annotation predominant. At several loci (including CDC123, ADCY5, KLHDC5) the combination of fine-mapping genetic data and chromatin state enrichment maps, supplemented by allelic imbalance in chromatin accessibility pinpointed likely causal variants. The combination of increasingly-precise genetic and islet epigenomic information accelerates definition of causal mechanisms implicated in T2D pathogenesis.

    View details for DOI 10.7554/eLife.31977

    View details for PubMedID 29412141

    View details for PubMedCentralID PMC5828664

  • Precision medicine in the management of type 2 diabetes. The lancet. Diabetes & endocrinology Gloyn, A. L., Drucker, D. J. 2018; 6 (11): 891–900

    Abstract

    The study of type 2 diabetes has been driven by advances in human genetics, epigenetics, biomarkers, mechanistic studies, and large clinical trials, enabling new insights into disease susceptibility, pathophysiology, progression, and development of complications. Simultaneously, several new drug classes with different mechanisms of action have been introduced over the past two decades, accompanied by data about cardiovascular safety and non-glycaemic outcomes. In this Review, we critically examine the progress and integration of this new science into clinical practice, and review opportunities for enabling the use of precision medicine in the diagnosis and treatment of type 2 diabetes. We contrast the success in delivering personalised medicine for monogenic diabetes with the greater challenge of providing a precision medicine approach for type 2 diabetes, highlighting gaps, limitations, and areas requiring further study.

    View details for DOI 10.1016/S2213-8587(18)30052-4

    View details for PubMedID 29699867

  • Data Descriptor: Sequence data and association statistics from 12,940 type 2 diabetes cases and controls SCIENTIFIC DATA Flannick, J., Fuchsberger, C., Mahajan, A., Teslovich, T. M., Agarwala, V., Gaulton, K. J., Caulkins, L., Koesterer, R., Ma, C., Moutsianas, L., McCarthy, D. J., Rivas, M. A., Perry, J. B., Sim, X., Blackwell, T. W., Robertson, N. R., Rayner, N., Cingolani, P., Locke, A. E., Tajes, J., Highland, H. M., Dupuis, J., Chines, P. S., Lindgren, C. M., Hartl, C., Jackson, A. U., Chen, H., Huyghe, J. R., De Bunt, M., Pearson, R. D., Kumar, A., Muller-Nurasyid, M., Grarup, N., Stringham, H. M., Gamazon, E. R., Lee, J., Chen, Y., Scott, R. A., Below, J. E., Chen, P., Huang, J., Go, M., Stitzel, M. L., Pasko, D., Parker, S. J., Varga, T. V., Green, T., Beer, N. L., Day-Williams, A. G., Ferreira, T., Fingerlin, T., Horikoshi, M., Hu, C., Huh, I., Ikram, M., Kim, B., Kim, Y., Kim, Y., Kwon, M., Lee, J., Lee, S., Lin, K., Maxwell, T. J., Nagai, Y., Wang, X., Welch, R. P., Yoon, J., Zhang, W., Barzilai, N., Voight, B. F., Han, B., Jenkinson, C. P., Kuulasmaa, T., Kuusisto, J., Manning, A., Ng, M. Y., Palmer, N. D., Balkau, B., Stancakova, A., Abboud, H. E., Boeing, H., Giedraitis, V., Prabhakaran, D., Gottesman, O., Scott, J., Carey, J., Kwan, P., Grant, G., Smith, J. D., Neale, B. M., Purcell, S., Butterworth, A. S., Howson, J. M., Lee, H., Lu, Y., Kwak, S., Zhao, W., Danesh, J., Lam, V. L., Park, K., Saleheen, D., So, W., Tam, C. T., Afzal, U., Aguilar, D., Arya, R., Aung, T., Chan, E., Navarro, C., Cheng, C., Palli, D., Correa, A., Curran, J. E., Rybin, D., Farook, V. S., Fowler, S. P., Freedman, B. I., Griswold, M., Hale, D., Hicks, P. J., Khor, C., Kumar, S., Lehne, B., Thuillier, D., Lim, W., Liu, J., Loh, M., Musani, S. K., Puppala, S., Scott, W. R., Yengo, L., Tan, S., Taylor, H. A., Thameem, F., Wilson, G., Wong, T., Njolstad, P., Levy, J. C., Mangino, M., Bonnycastle, L. L., Schwarzmayr, T., Fadista, J., Surdulescu, G. L., Herder, C., Groves, C. J., Wieland, T., Bork-Jensen, J., Brandslund, I., Christensen, C., Koistinen, H. A., Doney, A. F., Kinnunen, L., Esko, T., Farmer, A. J., Hakaste, L., Hodgkiss, D., Kravic, J., Lyssenko, V., Hollensted, M., Jorgensen, M. E., Jorgensen, T., Ladenvall, C., Justesen, J., Karajamaki, A., Kriebel, J., Rathmann, W., Lannfelt, L., Lauritzen, T., Narisu, N., Linneberg, A., Melander, O., Milani, L., Neville, M., Orho-Melander, M., Qi, L., Qi, Q., Roden, M., Rolandsson, O., Swift, A., Rosengren, A. H., Stirrups, K., Wood, A. R., Mihailov, E., Blancher, C., Carneiro, M. O., Maguire, J., Poplin, R., Shakir, K., Fennell, T., DePristo, M., De Angelis, M., Deloukas, P., Gjesing, A. P., Jun, G., Nilsson, P. M., Murphy, J., Onofrio, R., Thorand, B., Hansen, T., Meisinger, C., Hu, F. B., Isomaa, B., Karpe, F., Liang, L., Peters, A., Huth, C., O'Rahilly, S. P., Palmer, C. A., Pedersen, O., Rauramaa, R., Tuomilehto, J., Salomaa, V., Watanabe, R. M., Syvanen, A., Bergman, R. N., Bharadwaj, D., Bottinger, E. P., Cho, Y., Chandak, G. R., Chan, J., Chia, K., Daly, M. J., Ebrahim, S. B., Langenberg, C., Elliott, P., Jablonski, K. A., Lehman, D. M., Jia, W., Ma, R., Pollin, T. I., Sandhu, M., Tandon, N., Froguel, P., Barroso, I., Teo, Y., Zeggini, E., Loos, R. F., Small, K. S., Ried, J. S., DeFronzo, R. A., Grallert, H., Glaser, B., Metspalu, A., Wareham, N. J., Walker, M., Banks, E., Gieger, C., Ingelsson, E., Im, H., Illig, T., Franks, P. W., Buck, G., Trakalo, J., Buck, D., Prokopenko, I., Magi, R., Lind, L., Farjoun, Y., Owen, K. R., Gloyn, A. L., Strauch, K., Tuomi, T., Kooner, J., Lee, J., Park, T., Donnelly, P., Morris, A. D., Hattersley, A. T., Bowden, D. W., Collins, F. S., Atzmon, G., Chambers, J. C., Spector, T. D., Laakso, M., Strom, T. M., Bell, G. I., Blangero, J., Duggirala, R., Tai, E. S., McVean, G., Hanis, C. L., Wilson, J. G., Seielstad, M., Frayling, T. M., Meigs, J. B., Cox, N. J., Sladek, R., Lander, E. S., Gabriel, S., Mohlke, K. L., Meitinger, T., Groop, L., Abecasis, G., Scott, L. J., Morris, A. P., Kang, H., Altshuler, D., Burtt, N. P., Florez, J. C., Boehnke, M., McCarthy, M. I. 2017; 4: 170179

    Abstract

    To investigate the genetic basis of type 2 diabetes (T2D) to high resolution, the GoT2D and T2D-GENES consortia catalogued variation from whole-genome sequencing of 2,657 European individuals and exome sequencing of 12,940 individuals of multiple ancestries. Over 27M SNPs, indels, and structural variants were identified, including 99% of low-frequency (minor allele frequency [MAF] 0.1-5%) non-coding variants in the whole-genome sequenced individuals and 99.7% of low-frequency coding variants in the whole-exome sequenced individuals. Each variant was tested for association with T2D in the sequenced individuals, and, to increase power, most were tested in larger numbers of individuals (>80% of low-frequency coding variants in ~82 K Europeans via the exome chip, and ~90% of low-frequency non-coding variants in ~44 K Europeans via genotype imputation). The variants, genotypes, and association statistics from these analyses provide the largest reference to date of human genetic information relevant to T2D, for use in activities such as T2D-focused genotype imputation, functional characterization of variants or genes, and other novel analyses to detect associations between sequence variation and T2D.

    View details for PubMedID 29257133

  • Genes Associated with Pancreas Development and Function Maintain Open Chromatin in iPSCs Generated from Human Pancreatic Beta Cells STEM CELL REPORTS Thurner, M., Shenhav, L., Wesolowska-Andersen, A., Bennett, A. J., Barrett, A., Gloyn, A. L., McCarthy, M. I., Beer, N. L., Efrat, S. 2017; 9 (5): 1395–1405

    Abstract

    Current in vitro islet differentiation protocols suffer from heterogeneity and low efficiency. Induced pluripotent stem cells (iPSCs) derived from pancreatic beta cells (BiPSCs) preferentially differentiate toward endocrine pancreas-like cells versus those from fibroblasts (FiPSCs). We interrogated genome-wide open chromatin in BiPSCs and FiPSCs via ATAC-seq and identified ∼8.3k significant, differential open chromatin sites (DOCS) between the two iPSC subtypes (false discovery rate [FDR] < 0.05). DOCS where chromatin was more accessible in BiPSCs (Bi-DOCS) were significantly enriched for known regulators of endodermal development, including bivalent and weak enhancers, and FOXA2 binding sites (FDR < 0.05). Bi-DOCS were associated with genes related to pancreas development and beta-cell function, including transcription factors mutated in monogenic diabetes (PDX1, NKX2-2, HNF1A; FDR < 0.05). Moreover, Bi-DOCS correlated with enhanced gene expression in BiPSC-derived definitive endoderm and pancreatic progenitor cells. Bi-DOCS therefore highlight genes and pathways governing islet-lineage commitment, which can be exploited for differentiation protocol optimization, diabetes disease modeling, and therapeutic purposes.

    View details for DOI 10.1016/j.stemcr.2017.09.020

    View details for Web of Science ID 000415139400006

    View details for PubMedID 29107594

    View details for PubMedCentralID PMC5831005

  • Prioritising Causal Genes at Type 2 Diabetes Risk Loci CURRENT DIABETES REPORTS Grotz, A. K., Gloyn, A. L., Thomsen, S. K. 2017; 17 (9): 76

    Abstract

    Genome-wide association studies (GWAS) for type 2 diabetes (T2D) risk have identified a large number of genetic loci associated with disease susceptibility. However, progress moving from association signals through causal genes to functional understanding has so far been slow, hindering clinical translation. This review discusses the benefits and limitations of emerging, unbiased approaches for prioritising causal genes at T2D risk loci.Candidate causal genes can be identified by a number of different strategies that rely on genetic data, genomic annotations, and functional screening of selected genes. To overcome the limitations of each particular method, integration of multiple data sets is proving essential for establishing confidence in the prioritised genes. Previous studies have also highlighted the need to support these efforts through identification of causal variants and disease-relevant tissues. Prioritisation of causal genes at T2D risk loci by integrating complementary lines of evidence promises to accelerate our understanding of disease pathology and promote translation into new therapeutics.

    View details for DOI 10.1007/s11892-017-0907-y

    View details for Web of Science ID 000407447500011

    View details for PubMedID 28758174

    View details for PubMedCentralID PMC5534459

  • Human genetics as a model for target validation: finding new therapies for diabetes DIABETOLOGIA Thomsen, S. K., Gloyn, A. L. 2017; 60 (6): 960–70

    Abstract

    Type 2 diabetes is a global epidemic with major effects on healthcare expenditure and quality of life. Currently available treatments are inadequate for the prevention of comorbidities, yet progress towards new therapies remains slow. A major barrier is the insufficiency of traditional preclinical models for predicting drug efficacy and safety. Human genetics offers a complementary model to assess causal mechanisms for target validation. Genetic perturbations are 'experiments of nature' that provide a uniquely relevant window into the long-term effects of modulating specific targets. Here, we show that genetic discoveries over the past decades have accurately predicted (now known) therapeutic mechanisms for type 2 diabetes. These findings highlight the potential for use of human genetic variation for prospective target validation, and establish a framework for future applications. Studies into rare, monogenic forms of diabetes have also provided proof-of-principle for precision medicine, and the applicability of this paradigm to complex disease is discussed. Finally, we highlight some of the limitations that are relevant to the use of genome-wide association studies (GWAS) in the search for new therapies for diabetes. A key outstanding challenge is the translation of GWAS signals into disease biology and we outline possible solutions for tackling this experimental bottleneck.

    View details for DOI 10.1007/s00125-017-4270-y

    View details for Web of Science ID 000400995400003

    View details for PubMedID 28447115

    View details for PubMedCentralID PMC5423999

  • Variant Enriched in the Finnish Population is Associated With Fasting Insulin Levels and Type 2 Diabetes Risk. Diabetes Manning, A., Highland, H. M., Gasser, J., Sim, X., Tukiainen, T., Fontanillas, P., Grarup, N., Rivas, M. A., Mahajan, A., Locke, A. E., Cingolani, P., Pers, T. H., Viñuela, A., Brown, A. A., Wu, Y., Flannick, J., Fuchsberger, C., Gamazon, E. R., Gaulton, K. J., Im, H. K., Teslovich, T. M., Blackwell, T. W., Bork-Jensen, J., Burtt, N. P., Chen, Y., Green, T., Hartl, C., Kang, H. M., Kumar, A., Ladenvall, C., Ma, C., Moutsianas, L., Pearson, R. D., Perry, J. R., Rayner, N. W., Robertson, N. R., Scott, L. J., van de Bunt, M., Eriksson, J. G., Jula, A., Koskinen, S., Lehtimäki, T., Palotie, A., Raitakari, O. T., Jacobs, S. B., Wessel, J., Chu, A. Y., Scott, R. A., Goodarzi, M. O., Blancher, C., Buck, G., Buck, D., Chines, P. S., Gabriel, S., Gjesing, A. P., Groves, C. J., Hollensted, M., Huyghe, J. R., Jackson, A. U., Jun, G., Justesen, J. M., Mangino, M., Murphy, J., Neville, M., Onofrio, R., Small, K. S., Stringham, H. M., Trakalo, J., Banks, E., Carey, J., Carneiro, M. O., DePristo, M., Farjoun, Y., Fennell, T., Goldstein, J. I., Grant, G., Hrabé de Angelis, M., Maguire, J., Neale, B. M., Poplin, R., Purcell, S., Schwarzmayr, T., Shakir, K., Smith, J. D., Strom, T. M., Wieland, T., Lindstrom, J., Brandslund, I., Christensen, C., Surdulescu, G. L., Lakka, T. A., Doney, A. S., Nilsson, P., Wareham, N. J., Langenberg, C., Varga, T. V., Franks, P. W., Rolandsson, O., Rosengren, A. H., Farook, V. S., Thameem, F., Puppala, S., Kumar, S., Lehman, D. M., Jenkinson, C. P., Curran, J. E., Hale, D. E., Fowler, S. P., Arya, R., DeFronzo, R. A., Abboud, H. E., Syvänen, A., Hicks, P. J., Palmer, N. D., Ng, M. C., Bowden, D. W., Freedman, B. I., Esko, T., Mägi, R., Milani, L., Mihailov, E., Metspalu, A., Narisu, N., Kinnunen, L., Bonnycastle, L. L., Swift, A., Pasko, D., Wood, A. R., Fadista, J., Pollin, T. I., Barzilai, N., Atzmon, G., Glaser, B., Thorand, B., Strauch, K., Peters, A., Roden, M., Müller-Nurasyid, M., Liang, L., Kriebel, J., Illig, T., Grallert, H., Gieger, C., Meisinger, C., Lannfelt, L., Musani, S. K., Griswold, M., Taylor, H. A., Wilson, G., Correa, A., Oksa, H., Scott, W. R., Afzal, U., Tan, S., Loh, M., Chambers, J. C., Sehmi, J., Kooner, J. S., Lehne, B., Cho, Y. S., Lee, J., Han, B., Käräjämäki, A., Qi, Q., Qi, L., Huang, J., Hu, F. B., Melander, O., Orho-Melander, M., Below, J. E., Aguilar, D., Wong, T. Y., Liu, J., Khor, C., Chia, K. S., Lim, W. Y., Cheng, C., Chan, E., Tai, E. S., Aung, T., Linneberg, A., Isomaa, B., Meitinger, T., Tuomi, T., Hakaste, L., Kravic, J., Jørgensen, M. E., Lauritzen, T., Deloukas, P., Stirrups, K. E., Owen, K. R., Farmer, A. J., Frayling, T. M., O'Rahilly, S. P., Walker, M., Levy, J. C., Hodgkiss, D., Hattersley, A. T., Kuulasmaa, T., Stancáková, A., Barroso, I., Bharadwaj, D., Chan, J., Chandak, G. R., Daly, M. J., Donnelly, P. J., Ebrahim, S. B., Elliott, P., Fingerlin, T., Froguel, P., Hu, C., Jia, W., Ma, R. C., McVean, G., Park, T., Prabhakaran, D., Sandhu, M., Scott, J., Sladek, R., Tandon, N., Teo, Y. Y., Zeggini, E., Watanabe, R. M., Koistinen, H. A., Kesaniemi, Y. A., Uusitupa, M., Spector, T. D., Salomaa, V., Rauramaa, R., Palmer, C. N., Prokopenko, I., Morris, A. D., Bergman, R. N., Collins, F. S., Lind, L., Ingelsson, E., Tuomilehto, J., Karpe, F., Groop, L., Jørgensen, T., Hansen, T., Pedersen, O., Kuusisto, J., Abecasis, G., Bell, G. I., Blangero, J., Cox, N. J., Duggirala, R., Seielstad, M., Wilson, J. G., Dupuis, J., Ripatti, S., Hanis, C. L., Florez, J. C., Mohlke, K. L., Meigs, J. B., Laakso, M., Morris, A. P., Boehnke, M., Altshuler, D., McCarthy, M. I., Gloyn, A. L., Lindgren, C. M. 2017

    Abstract

    To identify novel coding association signals and facilitate characterization of mechanisms influencing glycemic traits and type 2 diabetes risk, we analyzed 109,215 variants derived from exome array genotyping together with an additional 390,225 variants from exome sequence in up to 39,339 normoglycemic individuals from five ancestry groups. We identified a novel association between the coding variant (p.Pro50Thr) in AKT2 and fasting plasma insulin (FI), a gene in which rare fully penetrant mutations are causal for monogenic glycemic disorders. The low-frequency allele is associated with a 12% increase in FI levels. This variant is present at 1.1% frequency in Finns but virtually absent in individuals from other ancestries. Carriers of the FI-increasing allele had increased 2-h insulin values, decreased insulin sensitivity, and increased risk of type 2 diabetes (odds ratio 1.05). In cellular studies, the AKT2-Thr50 protein exhibited a partial loss of function. We extend the allelic spectrum for coding variants in AKT2 associated with disorders of glucose homeostasis and demonstrate bidirectional effects of variants within the pleckstrin homology domain of AKT2.

    View details for DOI 10.2337/db16-1329

    View details for PubMedID 28341696

  • Decreased STARD10 Expression Is Associated with Defective Insulin Secretion in Humans and Mice AMERICAN JOURNAL OF HUMAN GENETICS Carrat, G. R., Hu, M., Nguyen-Tu, M., Chabosseau, P., Gaulton, K. J., van de Bunt, M., Siddiq, A., Falchi, M., Thurner, M., Canouil, M., Pattou, F., Leclerc, I., Pullen, T. J., Cane, M. C., Prabhala, P., Greenwald, W., Schulte, A., Marchetti, P., Ibberson, M., MacDonald, P. E., Fox, J., Gloyn, A. L., Froguel, P., Solimena, M., McCarthy, M. I., Rutter, G. A. 2017; 100 (2): 238–56

    Abstract

    Genetic variants near ARAP1 (CENTD2) and STARD10 influence type 2 diabetes (T2D) risk. The risk alleles impair glucose-induced insulin secretion and, paradoxically but characteristically, are associated with decreased proinsulin:insulin ratios, indicating improved proinsulin conversion. Neither the identity of the causal variants nor the gene(s) through which risk is conferred have been firmly established. Whereas ARAP1 encodes a GTPase activating protein, STARD10 is a member of the steroidogenic acute regulatory protein (StAR)-related lipid transfer protein family. By integrating genetic fine-mapping and epigenomic annotation data and performing promoter-reporter and chromatin conformational capture (3C) studies in β cell lines, we localize the causal variant(s) at this locus to a 5 kb region that overlaps a stretch-enhancer active in islets. This region contains several highly correlated T2D-risk variants, including the rs140130268 indel. Expression QTL analysis of islet transcriptomes from three independent subject groups demonstrated that T2D-risk allele carriers displayed reduced levels of STARD10 mRNA, with no concomitant change in ARAP1 mRNA levels. Correspondingly, β-cell-selective deletion of StarD10 in mice led to impaired glucose-stimulated Ca2+ dynamics and insulin secretion and recapitulated the pattern of improved proinsulin processing observed at the human GWAS signal. Conversely, overexpression of StarD10 in the adult β cell improved glucose tolerance in high fat-fed animals. In contrast, manipulation of Arap1 in β cells had no impact on insulin secretion or proinsulin conversion in mice. This convergence of human and murine data provides compelling evidence that the T2D risk associated with variation at this locus is mediated through reduction in STARD10 expression in the β cell.

    View details for DOI 10.1016/j.ajhg.2017.01.011

    View details for Web of Science ID 000393352000006

    View details for PubMedID 28132686

    View details for PubMedCentralID PMC5294761

  • The importance of Context: Uncovering Species- and Tissue-Specific Effects of Genetic Risk Variants for Type 2 Diabetes FRONTIERS IN ENDOCRINOLOGY Thomsen, S. K., McCarthy, M. I., Gloyn, A. L. 2016; 7: 112

    View details for DOI 10.3389/fendo.2016.00112

    View details for Web of Science ID 000385313400001

    View details for PubMedID 27630614

    View details for PubMedCentralID PMC5005446

  • The genetic architecture of type 2 diabetes NATURE Fuchsberger, C., Flannick, J., Teslovich, T. M., Mahajan, A., Agarwala, V., Gaulton, K. J., Ma, C., Fontanillas, P., Moutsianas, L., McCarthy, D. J., Rivas, M. A., Perry, J. R., Sim, X., Blackwell, T. W., Robertson, N. R., Rayner, N. W., Cingolani, P., Locke, A. E., Tajes, J. F., Highland, H. M., Dupuis, J., Chines, P. S., Lindgren, C. M., Hartl, C., Jackson, A. U., Chen, H., Huyghe, J. R., van de Bunt, M., Pearson, R. D., Kumar, A., Mueller-Nurasyid, M., Grarup, N., Stringham, H. M., Gamazon, E. R., Lee, J., Chen, Y., Scott, R. A., Below, J. E., Chen, P., Huang, J., Go, M. J., Stitzel, M. L., Pasko, D., Parker, S. C., Varga, T. V., Green, T., Beer, N. L., Day-Williams, A. G., Ferreira, T., Fingerlin, T., Horikoshi, M., Hu, C., Huh, I., Ikram, M. K., Kim, B., Kim, Y., Kim, Y. J., Kwon, M., Lee, J., Lee, S., Lin, K., Maxwell, T. J., Nagai, Y., Wang, X., Welch, R. P., Yoon, J., Zhang, W., Barzilai, N., Voight, B. F., Han, B., Jenkinson, C. P., Kuulasmaa, T., Kuusisto, J., Manning, A., Ng, M. C., Palmer, N. D., Balkau, B., Stancakova, A., Abboud, H. E., Boeing, H., Giedraitis, V., Prabhakaran, D., Gottesman, O., Scott, J., Carey, J., Kwan, P., Grant, G., Smith, J. D., Neale, B. M., Purcell, S., Butterworth, A. S., Howson, J. M., Lee, H. M., Lu, Y., Kwak, S., Zhao, W., Danesh, J., Lam, V. K., Park, K. S., Saleheen, D., So, W. Y., Tam, C. H., Afzal, U., Aguilar, D., Arya, R., Aung, T., Chan, E., Navarro, C., Cheng, C., Palli, D., Correa, A., Curran, J. E., Rybin, D., Farook, V. S., Fowler, S. P., Freedman, B. I., Griswold, M., Hale, D. E., Hicks, P. J., Khor, C., Kumar, S., Lehne, B., Thuillier, D., Lim, W. Y., Liu, J., van der Schouw, Y. T., Loh, M., Musani, S. K., Puppala, S., Scott, W. R., Yengo, L., Tan, S., Taylor, H. A., Thameem, F., Wilson, G., Wong, T. Y., Njolstad, P. R., Levy, J. C., Mangino, M., Bonnycastle, L. L., Schwarzmayr, T., Fadista, J., Surdulescu, G. L., Herder, C., Groves, C. J., Wieland, T., Bork-Jensen, J., Brandslund, I., Christensen, C., Koistinen, H. A., Doney, A. S., Kinnunen, L., Esko, T., Farmer, A. J., Hakaste, L., Hodgkiss, D., Kravic, J., Lyssenko, V., Hollensted, M., Jorgensen, M. E., Jorgensen, T., Ladenvall, C., Justesen, J. M., Karajamaki, A., Kriebel, J., Rathmann, W., Lannfelt, L., Lauritzen, T., Narisu, N., Linneberg, A., Melander, O., Milani, L., Neville, M., Orho-Melander, M., Qi, L., Qi, Q., Roden, M., Rolandsson, O., Swift, A., Rosengren, A. H., Stirrups, K., Wood, A. R., Mihailov, E., Blancher, C., Carneiro, M. O., Maguire, J., Poplin, R., Shakir, K., Fennell, T., DePristo, M., de Angelis, M. H., Deloukas, P., Gjesing, A. P., Jun, G., Nilsson, P., Murphy, J., Onofrio, R., Thorand, B., Hansen, T., Meisinger, C., Hu, F. B., Isomaa, B., Karpe, F., Liang, L., Peters, A., Huth, C., O'Rahilly, S. P., Palmer, C. N., Pedersen, O., Rauramaa, R., Tuomilehto, J., Salomaa, V., Watanabe, R. M., Syvanen, A., Bergman, R. N., Bharadwaj, D., Bottinger, E. P., Cho, Y. S., Chandak, G. R., Chan, J. C., Chia, K. S., Daly, M. J., Ebrahim, S. B., Langenberg, C., Elliott, P., Jablonski, K. A., Lehman, D. M., Jia, W., Ma, R. C., Pollin, T. I., Sandhu, M., Tandon, N., Froguel, P., Barroso, I., Teo, Y. Y., Zeggini, E., Loos, R. J., Small, K. S., Ried, J. S., DeFronzo, R. A., Grallert, H., Glaser, B., Metspalu, A., Wareham, N. J., Walker, M., Banks, E., Gieger, C., Ingelsson, E., Im, H. K., Illig, T., Franks, P. W., Buck, G., Trakalo, J., Buck, D., Prokopenko, I., Magi, R., Lind, L., Farjoun, Y., Owen, K. R., Gloyn, A. L., Strauch, K., Tuomi, T., Kooner, J. S., Lee, J., Park, T., Donnelly, P., Morris, A. D., Hattersley, A. T., Bowden, D. W., Collins, F. S., Atzmon, G., Chambers, J. C., Spector, T. D., Laakso, M., Strom, T. M., Bell, G. I., Blangero, J., Duggirala, R., Tai, E. S., McVean, G., Hanis, C. L., Wilson, J. G., Seielstad, M., Frayling, T. M., Meigs, J. B., Cox, N. J., Sladek, R., Lander, E. S., Gabriel, S., Burtt, N. P., Mohlke, K. L., Meitinger, T., Groop, L., Abecasis, G., Florez, J. C., Scott, L. J., Morris, A. P., Kang, H. M., Boehnke, M., Altshuler, D., McCarthy, M. I. 2016; 536 (7614): 41-?

    Abstract

    The genetic architecture of common traits, including the number, frequency, and effect sizes of inherited variants that contribute to individual risk, has been long debated. Genome-wide association studies have identified scores of common variants associated with type 2 diabetes, but in aggregate, these explain only a fraction of the heritability of this disease. Here, to test the hypothesis that lower-frequency variants explain much of the remainder, the GoT2D and T2D-GENES consortia performed whole-genome sequencing in 2,657 European individuals with and without diabetes, and exome sequencing in 12,940 individuals from five ancestry groups. To increase statistical power, we expanded the sample size via genotyping and imputation in a further 111,548 subjects. Variants associated with type 2 diabetes after sequencing were overwhelmingly common and most fell within regions previously identified by genome-wide association studies. Comprehensive enumeration of sequence variation is necessary to identify functional alleles that provide important clues to disease pathophysiology, but large-scale sequencing does not support the idea that lower-frequency variants have a major role in predisposition to type 2 diabetes.

    View details for DOI 10.1038/nature18642

    View details for PubMedID 27398621

  • Insights into metabolic disease from studying genetics in isolated populations: stories from Greece to Greenland DIABETOLOGIA Zeggini, E., Gloyn, A. L., Hansen, T. 2016; 59 (5): 938–41

    Abstract

    Over the last 10 years substantial progress has been made in our understanding of the genetic basis for type 2 diabetes and related traits. These developments have been facilitated by technological advancements that have allowed comprehensive genome-wide assessments of the impact of common genetic variation on disease risk. Current efforts are now focused on extending this to genetic variants in the rare and low-frequency spectrum by capitalising on next-generation sequencing technologies. This review discusses the important contributions that studies in isolated populations are making to this effort for diabetes and metabolic disease, drawing on specific examples from populations in Greece and Greenland. This review summarises a presentation given at the 'Exciting news in genetics of diabetes' symposium at the 2015 annual meeting of the EASD, with topics presented by Eleftheria Zeggini and Torben Hansen, and an overview by the Session Chair, Anna Gloyn.

    View details for DOI 10.1007/s00125-016-3926-3

    View details for Web of Science ID 000373993300008

    View details for PubMedID 26993633

    View details for PubMedCentralID PMC4826421

  • Systematic Functional Characterization of Candidate Causal Genes for Type 2 Diabetes Risk Variants. Diabetes Thomsen, S. K., Ceroni, A. n., van de Bunt, M. n., Burrows, C. n., Barrett, A. n., Scharfmann, R. n., Ebner, D. n., McCarthy, M. I., Gloyn, A. L. 2016; 65 (12): 3805–11

    Abstract

    Most genetic association signals for type 2 diabetes risk are located in noncoding regions of the genome, hindering translation into molecular mechanisms. Physiological studies have shown a majority of disease-associated variants to exert their effects through pancreatic islet dysfunction. Systematically characterizing the role of regional transcripts in β-cell function could identify the underlying disease-causing genes, but large-scale studies in human cellular models have previously been impractical. We developed a robust and scalable strategy based on arrayed gene silencing in the human β-cell line EndoC-βH1. In a screen of 300 positional candidates selected from 75 type 2 diabetes regions, each gene was assayed for effects on multiple disease-relevant phenotypes, including insulin secretion and cellular proliferation. We identified a total of 45 genes involved in β-cell function, pointing to possible causal mechanisms at 37 disease-associated loci. The results showed a strong enrichment for genes implicated in monogenic diabetes. Selected effects were validated in a follow-up study, including several genes (ARL15, ZMIZ1, and THADA) with previously unknown or poorly described roles in β-cell biology. We have demonstrated the feasibility of systematic functional screening in a human β-cell model and successfully prioritized plausible disease-causing genes at more than half of the regions investigated.

    View details for DOI 10.2337/db16-0361

    View details for PubMedID 27554474

    View details for PubMedCentralID PMC5402869

  • Insights into islet development and biology through characterization of a human iPSC-derived endocrine pancreas model ISLETS van de Bunt, M., Lako, M., Barrett, A., Gloyn, A. L., Hansson, M., McCarthy, M. I., Beer, N. L., Honore, C. 2016; 8 (3): 83–95

    Abstract

    Directed differentiation of stem cells offers a scalable solution to the need for human cell models recapitulating islet biology and T2D pathogenesis. We profiled mRNA expression at 6 stages of an induced pluripotent stem cell (iPSC) model of endocrine pancreas development from 2 donors, and characterized the distinct transcriptomic profiles associated with each stage. Established regulators of endodermal lineage commitment, such as SOX17 (log2 fold change [FC] compared to iPSCs = 14.2, p-value = 4.9 × 10(-5)) and the pancreatic agenesis gene GATA6 (log2 FC = 12.1, p-value = 8.6 × 10(-5)), showed transcriptional variation consistent with their known developmental roles. However, these analyses highlighted many other genes with stage-specific expression patterns, some of which may be novel drivers or markers of islet development. For example, the leptin receptor gene, LEPR, was most highly expressed in published data from in vivo-matured cells compared to our endocrine pancreas-like cells (log2 FC = 5.5, p-value = 2.0 × 10(-12)), suggesting a role for the leptin pathway in the maturation process. Endocrine pancreas-like cells showed significant stage-selective expression of adult islet genes, including INS, ABCC8, and GLP1R, and enrichment of relevant GO-terms (e.g. "insulin secretion"; odds ratio = 4.2, p-value = 1.9 × 10(-3)): however, principal component analysis indicated that in vitro-differentiated cells were more immature than adult islets. Integration of the stage-specific expression information with genetic data from T2D genome-wide association studies revealed that 46 of 82 T2D-associated loci harbor genes present in at least one developmental stage, facilitating refinement of potential effector transcripts. Together, these data show that expression profiling in an iPSC islet development model can further understanding of islet biology and T2D pathogenesis.

    View details for DOI 10.1080/19382014.2016.1182276

    View details for Web of Science ID 000377815900003

    View details for PubMedID 27246810

    View details for PubMedCentralID PMC4987020

  • Loss-of-Function Mutations in the Cell-Cycle Control Gene CDKN2A Impact on Glucose Homeostasis in Humans. Diabetes Pal, A. n., Potjer, T. P., Thomsen, S. K., Ng, H. J., Barrett, A. n., Scharfmann, R. n., James, T. J., Bishop, D. T., Karpe, F. n., Godsland, I. F., Vasen, H. F., Newton-Bishop, J. n., Pijl, H. n., McCarthy, M. I., Gloyn, A. L. 2016; 65 (2): 527–33

    Abstract

    At the CDKN2A/B locus, three independent signals for type 2 diabetes risk are located in a noncoding region near CDKN2A. The disease-associated alleles have been implicated in reduced β-cell function, but the underlying mechanism remains elusive. In mice, β-cell-specific loss of Cdkn2a causes hyperplasia, while overexpression leads to diabetes, highlighting CDKN2A as a candidate effector transcript. Rare CDKN2A loss-of-function mutations are a cause of familial melanoma and offer the opportunity to determine the impact of CDKN2A haploinsufficiency on glucose homeostasis in humans. To test the hypothesis that such individuals have improved β-cell function, we performed oral and intravenous glucose tolerance tests on mutation carriers and matched control subjects. Compared with control subjects, carriers displayed increased insulin secretion, impaired insulin sensitivity, and reduced hepatic insulin clearance. These results are consistent with a model whereby CDKN2A loss affects a range of different tissues, including pancreatic β-cells and liver. To test for direct effects of CDKN2A-loss on β-cell function, we performed knockdown in a human β-cell line, EndoC-bH1. This revealed increased insulin secretion independent of proliferation. Overall, we demonstrated that CDKN2A is an important regulator of glucose homeostasis in humans, thus supporting its candidacy as an effector transcript for type 2 diabetes-associated alleles in the region.

    View details for DOI 10.2337/db15-0602

    View details for PubMedID 26542317

    View details for PubMedCentralID PMC4724950

  • Genome-edited human stem cell-derived beta cells: a powerful tool for drilling down on type 2 diabetes GWAS biology. F1000Research Beer, N. L., Gloyn, A. L. 2016; 5

    Abstract

    Type 2 diabetes (T2D) is a disease of pandemic proportions, one defined by a complex aetiological mix of genetic, epigenetic, environmental, and lifestyle risk factors. Whilst the last decade of T2D genetic research has identified more than 100 loci showing strong statistical association with disease susceptibility, our inability to capitalise upon these signals reflects, in part, a lack of appropriate human cell models for study. This review discusses the impact of two complementary, state-of-the-art technologies on T2D genetic research: the generation of stem cell-derived, endocrine pancreas-lineage cells and the editing of their genomes. Such models facilitate investigation of diabetes-associated genomic perturbations in a physiologically representative cell context and allow the role of both developmental and adult islet dysfunction in T2D pathogenesis to be investigated. Accordingly, we interrogate the role that patient-derived induced pluripotent stem cell models are playing in understanding cellular dysfunction in monogenic diabetes, and how site-specific nucleases such as the clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 system are helping to confirm genes crucial to human endocrine pancreas development. We also highlight the novel biology gleaned in the absence of patient lines, including an ability to model the whole phenotypic spectrum of diabetes phenotypes occurring both in utero and in adult cells, interrogating the non-coding 'islet regulome' for disease-causing perturbations, and understanding the role of other islet cell types in aberrant glycaemia. This article aims to reinforce the importance of investigating T2D signals in cell models reflecting appropriate species, genomic context, developmental time point, and tissue type.

    View details for DOI 10.12688/f1000research.8682.1

    View details for PubMedID 27508066

    View details for PubMedCentralID PMC4955023

  • Transcript Expression Data from Human Islets Links Regulatory Signals from Genome-Wide Association Studies for Type 2 Diabetes and Glycemic Traits to Their Downstream Effectors PLOS GENETICS van de Bunt, M., Fox, J., Dai, X., Barrett, A., Grey, C., Li, L., Bennett, A. J., Johnson, P. R., Rajotte, R. V., Gaulton, K. J., Dermitzakis, E. T., MacDonald, P. E., McCarthy, M. I., Gloyn, A. L. 2015; 11 (12): e1005694

    Abstract

    The intersection of genome-wide association analyses with physiological and functional data indicates that variants regulating islet gene transcription influence type 2 diabetes (T2D) predisposition and glucose homeostasis. However, the specific genes through which these regulatory variants act remain poorly characterized. We generated expression quantitative trait locus (eQTL) data in 118 human islet samples using RNA-sequencing and high-density genotyping. We identified fourteen loci at which cis-exon-eQTL signals overlapped active islet chromatin signatures and were coincident with established T2D and/or glycemic trait associations. ‎At some, these data provide an experimental link between GWAS signals and biological candidates, such as DGKB and ADCY5. At others, the cis-signals implicate genes with no prior connection to islet biology, including WARS and ZMIZ1. At the ZMIZ1 locus, we show that perturbation of ZMIZ1 expression in human islets and beta-cells influences exocytosis and insulin secretion, highlighting a novel role for ZMIZ1 in the maintenance of glucose homeostasis. Together, these findings provide a significant advance in the mechanistic insights of T2D and glycemic trait association loci.

    View details for PubMedID 26624892

  • Genetic fine mapping and genomic annotation defines causal mechanisms at type 2 diabetes susceptibility loci NATURE GENETICS Gaulton, K. J., Ferreira, T., Lee, Y., Raimondo, A., Maegi, R., Reschen, M. E., Mahajan, A., Locke, A., Rayner, N. W., Robertson, N., Scott, R. A., Prokopenko, I., Scott, L. J., Green, T., Sparso, T., Thuillier, D., Yengo, L., Grallert, H., Wahl, S., Franberg, M., Strawbridge, R. J., Kestler, H., Chheda, H., Eisele, L., Gustafsson, S., Steinthorsdottir, V., Thorleifsson, G., Qi, L., Karssen, L. C., van Leeuwen, E. M., Willems, S. M., Li, M., Chen, H., Fuchsberger, C., Kwan, P., Ma, C., Linderman, M., Lu, Y., Thomsen, S. K., Rundle, J. K., Beer, N. L., van de Bunt, M., Chalisey, A., Kang, H. M., Voight, B. F., Abecasis, G. R., Almgren, P., Baldassarre, D., Balkau, B., Benediktsson, R., Blueher, M., Boeing, H., Bonnycastle, L. L., Bottinger, E. P., Burtt, N. P., Carey, J., Charpentier, G., Chines, P. S., Cornelis, M. C., Couper, D. J., Crenshaw, A. T., van Dam, R. M., Doney, A. S., Dorkhan, M., Edkins, S., Eriksson, J. G., Esko, T., Eury, E., Fadista, J., Flannick, J., Fontanillas, P., Fox, C., Franks, P. W., Gertow, K., Gieger, C., Gigante, B., Gottesman, O., Grant, G. B., Grarup, N., Groves, C. J., Hassinen, M., Have, C. T., Herder, C., Holmen, O. L., Hreidarsson, A. B., Humphries, S. E., Hunter, D. J., Jackson, A. U., Jonsson, A., Jorgensen, M. E., Jorgensen, T., Kao, W. L., Kerrison, N. D., Kinnunen, L., Klopp, N., Kong, A., Kovacs, P., Kraft, P., Kravic, J., Langford, C., Leander, K., Liang, L., Lichtner, P., Lindgren, C. M., Lindholm, E., Linneberg, A., Liu, C., Lobbens, S., Luan, J., Lyssenko, V., Mannisto, S., McLeod, O., Meyer, J., Mihailov, E., Mirza, G., Muehleisen, T. W., Mueller-Nurasyid, M., Navarro, C., Noethen, M. M., Oskolkov, N. N., Owen, K. R., Palli, D., Pechlivanis, S., Peltonen, L., Perry, J. R., Platou, C. G., Roden, M., Ruderfer, D., Rybin, D., van der Schouw, Y. T., Sennblad, B., Sigurdsson, G., Stancakova, A., Steinbach, G., Storm, P., Strauch, K., Stringham, H. M., Sun, Q., Thorand, B., Tikkanen, E., Tonjes, A., Trakalo, J., Tremoli, E., Tuomi, T., Wennauer, R., Wiltshire, S., Wood, A. R., Zeggini, E., Dunham, I., Birney, E., Pasquali, L., Ferrer, J., Loos, R. J., Dupuis, J., Florez, J. C., Boerwinkle, E., Pankow, J. S., Van Duijn, C., Sijbrands, E., Meigs, J. B., Hu, F. B., Thorsteinsdottir, U., Stefansson, K., Lakka, T. A., Rauramaa, R., Stumvoll, M., Pedersen, N. L., Lind, L., Keinanen-Kiukaanniemi, S. M., Korpi-Hyovalti, E., Saaristo, T. E., Saltevo, J., Kuusisto, J., Laakso, M., Metspalu, A., Erbel, R., Joecke, K., Moebus, S., Ripatti, S., Salomaa, V., Ingelsson, E., Boehm, B. O., Bergman, R. N., Collins, F. S., Mohlke, K. L., Koistinen, H., Tuomilehto, J., Hveem, K., Njolstad, I., Deloukas, P., Donnelly, P. J., Frayling, T. M., Hattersley, A. T., de Faire, U., Hamsten, A., Illig, T., Peters, A., Cauchi, S., Sladek, R., Froguel, P., Hansen, T., Pedersen, O., Morris, A. D., Palmer, C. N., Kathiresan, S., Melander, O., Nilsson, P. M., Groop, L. C., Barroso, I., Langenberg, C., Wareham, N. J., O'Callaghan, C. A., Gloyn, A. L., Altshuler, D., Boehnke, M., Teslovich, T. M., McCarthy, M. I., Morris, A. P. 2015; 47 (12): 1415-?

    Abstract

    We performed fine mapping of 39 established type 2 diabetes (T2D) loci in 27,206 cases and 57,574 controls of European ancestry. We identified 49 distinct association signals at these loci, including five mapping in or near KCNQ1. 'Credible sets' of the variants most likely to drive each distinct signal mapped predominantly to noncoding sequence, implying that association with T2D is mediated through gene regulation. Credible set variants were enriched for overlap with FOXA2 chromatin immunoprecipitation binding sites in human islet and liver cells, including at MTNR1B, where fine mapping implicated rs10830963 as driving T2D association. We confirmed that the T2D risk allele for this SNP increases FOXA2-bound enhancer activity in islet- and liver-derived cells. We observed allele-specific differences in NEUROD1 binding in islet-derived cells, consistent with evidence that the T2D risk allele increases islet MTNR1B expression. Our study demonstrates how integration of genetic and genomic information can define molecular mechanisms through which variants underlying association signals exert their effects on disease.

    View details for DOI 10.1038/ng.3437

    View details for PubMedID 26551672

  • Isocitrate-to-SENP1 signaling amplifies insulin secretion and rescues dysfunctional beta cells JOURNAL OF CLINICAL INVESTIGATION Ferdaoussi, M., Dai, X., Jensen, M. V., Wang, R., Peterson, B. S., Huang, C., Ilkayeva, O., Smith, N., Miller, N., Hajmrle, C., Spigelman, A. F., Wright, R. C., Plummer, G., Suzuki, K., Mackay, J. P., van de Bunt, M., Gloyn, A. L., Ryan, T. E., Norquay, L. D., Brosnan, M., Trimmer, J. K., Rolph, T. P., Kibbey, R. G., Fox, J., Colmers, W. F., Shirihai, O. S., Neufer, P., Yeh, E. H., Newgard, C. B., MacDonald, P. E. 2015; 125 (10): 3847–60

    Abstract

    Insulin secretion from β cells of the pancreatic islets of Langerhans controls metabolic homeostasis and is impaired in individuals with type 2 diabetes (T2D). Increases in blood glucose trigger insulin release by closing ATP-sensitive K+ channels, depolarizing β cells, and opening voltage-dependent Ca2+ channels to elicit insulin exocytosis. However, one or more additional pathway(s) amplify the secretory response, likely at the distal exocytotic site. The mitochondrial export of isocitrate and engagement with cytosolic isocitrate dehydrogenase (ICDc) may be one key pathway, but the mechanism linking this to insulin secretion and its role in T2D have not been defined. Here, we show that the ICDc-dependent generation of NADPH and subsequent glutathione (GSH) reduction contribute to the amplification of insulin exocytosis via sentrin/SUMO-specific protease-1 (SENP1). In human T2D and an in vitro model of human islet dysfunction, the glucose-dependent amplification of exocytosis was impaired and could be rescued by introduction of signaling intermediates from this pathway. Moreover, islet-specific Senp1 deletion in mice caused impaired glucose tolerance by reducing the amplification of insulin exocytosis. Together, our results identify a pathway that links glucose metabolism to the amplification of insulin secretion and demonstrate that restoration of this axis rescues β cell function in T2D.

    View details for DOI 10.1172/JCI82498

    View details for Web of Science ID 000362311700016

    View details for PubMedID 26389676

    View details for PubMedCentralID PMC4607115

  • When is it MODY? Challenges in the Interpretation of Sequence Variants in MODY Genes. The review of diabetic studies : RDS Althari, S., Gloyn, A. L. 2015; 12 (3-4): 330–48

    Abstract

    The genomics revolution has raised more questions than it has provided answers. Big data from large population-scale resequencing studies are increasingly deconstructing classic notions of Mendelian disease genetics, which support a simplistic correlation between mutational severity and phenotypic outcome. The boundaries are being blurred as the body of evidence showing monogenic disease-causing alleles in healthy genomes, and in the genomes of individu-als with increased common complex disease risk, continues to grow. In this review, we focus on the newly emerging challenges which pertain to the interpretation of sequence variants in genes implicated in the pathogenesis of maturity-onset diabetes of the young (MODY), a presumed mono-genic form of diabetes characterized by Mendelian inheritance. These challenges highlight the complexities surrounding the assignments of pathogenicity, in particular to rare protein-alerting variants, and bring to the forefront some profound clinical diagnostic implications. As MODY is both genetically and clinically heterogeneous, an accurate molecular diagnosis and cautious extrapolation of sequence data are critical to effective disease management and treatment. The biological and translational value of sequence information can only be attained by adopting a multitude of confirmatory analyses, which interrogate variant implication in disease from every possible angle. Indeed, studies which have effectively detected rare damaging variants in known MODY genes in normoglycemic individuals question the existence of a sin-gle gene mutation scenario: does monogenic diabetes exist when the genetic culprits of MODY have been systematical-ly identified in individuals without MODY?

    View details for DOI 10.1900/RDS.2015.12.330

    View details for PubMedID 27111119

  • Human islet function following 20 years of cryogenic biobanking DIABETOLOGIA Fox, J., Lyon, J., Dai, X., Wright, R. C., Hayward, J., van de Bunt, M., Kin, T., Shapiro, A., McCarthy, M. I., Gloyn, A. L., Ungrin, M. D., Lakey, J. R., Kneteman, N. M., Warnock, G. L., Korbutt, G. S., Rajotte, R. V., MacDonald, P. E. 2015; 58 (7): 1503–12

    Abstract

    There are potential advantages to the low-temperature (-196 °C) banking of isolated islets, including the maintenance of viable islets for future research. We therefore assessed the in vitro and in vivo function of islets cryopreserved for nearly 20 years.Human islets were cryopreserved from 1991 to 2001 and thawed between 2012 and 2014. These were characterised by immunostaining, patch-clamp electrophysiology, insulin secretion, transcriptome analysis and transplantation into a streptozotocin (STZ)-induced mouse model of diabetes.The cryopreservation time was 17.6 ± 0.4 years (n = 43). The thawed islets stained positive with dithizone, contained insulin-positive and glucagon-positive cells, and displayed levels of apoptosis and transcriptome profiles similar to those of freshly isolated islets, although their insulin content was lower. The cryopreserved beta cells possessed ion channels and exocytotic responses identical to those of freshly isolated beta cells. Cells from a subset of five donors demonstrated similar perifusion insulin secretion profiles pre- and post-cryopreservation. The transplantation of cryopreserved islets into the diabetic mice improved their glucose tolerance but did not completely normalise their blood glucose levels. Circulating human insulin and insulin-positive grafts were detectable at 10 weeks post-transplantation.We have demonstrated the potential for long-term banking of human islets for research, which could enable the use of tissue from a large number of donors with future technologies to gain new insight into diabetes.

    View details for DOI 10.1007/s00125-015-3598-4

    View details for Web of Science ID 000356528900015

    View details for PubMedID 25930156

    View details for PubMedCentralID PMC4472956

  • Recognition and Management of Individuals With Hyperglycemia Because of a Heterozygous Glucokinase Mutation DIABETES CARE Chakera, A. J., Steele, A. M., Gloyn, A. L., Shepherd, M. H., Shields, B., Ellard, S., Hattersley, A. T. 2015; 38 (7): 1383–92

    Abstract

    Glucokinase-maturity-onset diabetes of the young (GCK-MODY), also known as MODY2, is caused by heterozygous inactivating mutations in the GCK gene. GCK gene mutations are present in ∼1 in 1,000 of the population, but most are not diagnosed. They are common causes of MODY (10-60%): persistent incidental childhood hyperglycemia (10-60%) and gestational diabetes mellitus (1-2%). GCK-MODY has a unique pathophysiology and clinical characteristics, so it is best considered as a discrete genetic subgroup. People with GCK-MODY have a defect in glucose sensing; hence, glucose homeostasis is maintained at a higher set point resulting in mild, asymptomatic fasting hyperglycemia (5.4-8.3 mmol/L, HbA1c range 5.8-7.6% [40-60 mmol/mol]), which is present from birth and shows slight deterioration with age. Even after 50 years of mild hyperglycemia, people with GCK-MODY do not develop significant microvascular complications, and the prevalence of macrovascular complications is probably similar to that in the general population. Treatment is not recommended outside pregnancy because glucose-lowering therapy is ineffective in people with GCK-MODY and there is a lack of long-term complications. In pregnancy, fetal growth is primarily determined by whether the fetus inherits the GCK gene mutation from their mother. Insulin treatment of the mother is only appropriate when increased fetal abdominal growth on scanning suggests the fetus is unaffected. The impact on outcome of maternal insulin treatment is limited owing to the difficulty in altering maternal glycemia in these patients. Making the diagnosis of GCK-MODY through genetic testing is essential to avoid unnecessary treatment and investigations, especially when patients are misdiagnosed with type 1 or type 2 diabetes.

    View details for DOI 10.2337/dc14-2769

    View details for Web of Science ID 000356933600035

    View details for PubMedID 26106223

  • Glucokinase regulatory protein: complexity at the crossroads of triglyceride and glucose metabolism CURRENT OPINION IN LIPIDOLOGY Raimondo, A., Rees, M. G., Gloyn, A. L. 2015; 26 (2): 88–95

    Abstract

    Glucokinase regulator (GCKR) encodes glucokinase regulatory protein (GKRP), a hepatocyte-specific inhibitor of the glucose-metabolizing enzyme glucokinase (GCK). Genome-wide association studies have identified a common coding variant within GCKR associated with multiple metabolic traits. This review focuses on recent insights into the critical role of GKRP in hepatic glucose metabolism that have stemmed from the study of human genetics. This knowledge has improved our understanding of glucose and lipid physiology and informed the development of targeted molecular therapeutics for diabetes.Rare GCKR variants have effects on GKRP expression, localization, and activity. These variants are collectively associated with hypertriglyceridaemia but are not causal. Crystal structures of GKRP and the GCK-GKRP complex have been solved, providing greater insight into the molecular interactions between these proteins. Finally, small molecules have been identified that directly bind GKRP and reduce blood glucose levels in rodent models of diabetes.GCKR variants across the allelic spectrum have effects on glucose and lipid homeostasis. Functional analysis has highlighted numerous molecular mechanisms for GKRP dysfunction. Hepatocyte-specific GCK activation via small molecule GKRP inhibition may be a new avenue for type 2 diabetes treatment, particularly considering evidence indicating GKRP loss-of-function alone does not cause hypertriglyceridaemia.

    View details for DOI 10.1097/MOL.0000000000000155

    View details for Web of Science ID 000352227500004

    View details for PubMedID 25692341

    View details for PubMedCentralID PMC4422901

  • Identification and Functional Characterization of G6PC2 Coding Variants Influencing Glycemic Traits Define an Effector Transcript at the G6PC2-ABCB11 Locus PLOS GENETICS Mahajan, A., Sim, X., Ng, H. J., Manning, A., Rivas, M. A., Highland, H. M., Locke, A. E., Grarup, N., Im, H. K., Cingolani, P., Flannick, J., Fontanillas, P., Fuchsberger, C., Gaulton, K. J., Teslovich, T. M., Rayner, N. W., Robertson, N. R., Beer, N. L., Rundle, J. K., Bork-Jensen, J., Ladenvall, C., Blancher, C., Buck, D., Buck, G., Burtt, N. P., Gabriel, S., Gjesing, A. P., Groves, C. J., Hollensted, M., Huyghe, J. R., Jackson, A. U., Jun, G., Justesen, J. M., Mangino, M., Murphy, J., Neville, M., Onofrio, R., Small, K. S., Stringham, H. M., Syvanen, A., Trakalo, J., Abecasis, G., Bell, G. I., Blangero, J., Cox, N. J., Duggirala, R., Hanis, C. L., Seielstad, M., Wilson, J. G., Christensen, C., Brandslund, I., Rauramaa, R., Surdulescu, G. L., Doney, A. S., Lannfelt, L., Linneberg, A., Isomaa, B., Tuomi, T., Jorgensen, M. E., Jorgensen, T., Kuusisto, J., Uusitupa, M., Salomaa, V., Spector, T. D., Morris, A. D., Palmer, C. N., Collins, F. S., Mohlke, K. L., Bergman, R. N., Ingelsson, E., Lind, L., Tuomilehto, J., Hansen, T., Watanabe, R. M., Prokopenko, I., Dupuis, J., Karpe, F., Groop, L., Laakso, M., Pedersen, O., Florez, J. C., Morris, A. P., Altshuler, D., Meigs, J. B., Boehnke, M., McCarthy, M. I., Lindgren, C. M., Gloyn, A. L. 2015; 11 (1)

    Abstract

    Genome wide association studies (GWAS) for fasting glucose (FG) and insulin (FI) have identified common variant signals which explain 4.8% and 1.2% of trait variance, respectively. It is hypothesized that low-frequency and rare variants could contribute substantially to unexplained genetic variance. To test this, we analyzed exome-array data from up to 33,231 non-diabetic individuals of European ancestry. We found exome-wide significant (P<5×10-7) evidence for two loci not previously highlighted by common variant GWAS: GLP1R (p.Ala316Thr, minor allele frequency (MAF)=1.5%) influencing FG levels, and URB2 (p.Glu594Val, MAF = 0.1%) influencing FI levels. Coding variant associations can highlight potential effector genes at (non-coding) GWAS signals. At the G6PC2/ABCB11 locus, we identified multiple coding variants in G6PC2 (p.Val219Leu, p.His177Tyr, and p.Tyr207Ser) influencing FG levels, conditionally independent of each other and the non-coding GWAS signal. In vitro assays demonstrate that these associated coding alleles result in reduced protein abundance via proteasomal degradation, establishing G6PC2 as an effector gene at this locus. Reconciliation of single-variant associations and functional effects was only possible when haplotype phase was considered. In contrast to earlier reports suggesting that, paradoxically, glucose-raising alleles at this locus are protective against type 2 diabetes (T2D), the p.Val219Leu G6PC2 variant displayed a modest but directionally consistent association with T2D risk. Coding variant associations for glycemic traits in GWAS signals highlight PCSK1, RREB1, and ZHX3 as likely effector transcripts. These coding variant association signals do not have a major impact on the trait variance explained, but they do provide valuable biological insights.

    View details for DOI 10.1371/journal.pgen.1004876

    View details for Web of Science ID 000349314600012

    View details for PubMedID 25625282

    View details for PubMedCentralID PMC4307976

  • The pancreatic beta cell: recent insights from human genetics TRENDS IN ENDOCRINOLOGY AND METABOLISM Thomsen, S. K., Gloyn, A. L. 2014; 25 (8): 425–34

    Abstract

    Diabetes mellitus is a metabolic disease characterised by relative or absolute pancreatic β cell dysfunction. Genetic variants implicated in disease risk can be identified by studying affected individuals. To understand the mechanisms driving genetic associations, variants must be translated through causative transcripts to biological insights. Studies into the genetic basis of Mendelian forms of diabetes have successfully identified genes involved in both β cell function and pancreatic development. For type 2 diabetes (T2D), genome-wide association studies (GWASs) are uncovering an ever-increasing number of susceptibility variants that exert their effect through β cell dysfunction, but translation to mechanistic understanding has in most cases been slow. Improved annotations of the islet genome and advances in whole-genome and -exome sequencing (WHS and WES) have facilitated recent progress.

    View details for DOI 10.1016/j.tem.2014.05.001

    View details for Web of Science ID 000340312900008

    View details for PubMedID 24986330

    View details for PubMedCentralID PMC4229643

  • Reclassification of Diabetes Etiology in a Family With Multiple Diabetes Phenotypes JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM Kavvoura, F. K., Raimondo, A., Thanabalasingham, G., Barrett, A., Webster, A. L., Shears, D., Mann, N. P., Ellard, S., Gloyn, A. L., Owen, K. R. 2014; 99 (6): E1067–E1071

    Abstract

    Maturity-onset diabetes of the young (MODY) is uncommon; however, accurate diagnosis facilitates personalized management and informs prognosis in probands and relatives.The objective of the study was to highlight that the appropriate use of genetic and nongenetic investigations leads to the correct classification of diabetes etiology.A 30-year-old European female was diagnosed with insulin-treated gestational diabetes. She discontinued insulin after delivery; however, her fasting hyperglycemia persisted. β-Cell antibodies were negative and C-peptide was 0.79 nmol/L. Glucokinase (GCK)-MODY was suspected and confirmed by the identification of a GCK mutation (p.T206M).Systematic clinical and biochemical characterization and GCK mutational analysis were implemented to determine the diabetes etiology in five relatives. Functional characterization of GCK mutations was performed.Identification of the p.T206M mutation in the proband's sister confirmed a diagnosis of GCK-MODY. Her daughter was diagnosed at 16 weeks with permanent neonatal diabetes (PNDM). Mutation analysis identified two GCK mutations that were inherited in trans-p. [(R43P);(T206M)], confirming a diagnosis of GCK-PNDM. Both mutations were shown to be kinetically inactivating. The proband's mother, other sister, and daughter all had a clinical diagnosis of type 1 diabetes, confirmed by undetectable C-peptide levels and β-cell antibody positivity. GCK mutations were not detected.Two previously misclassified family members were shown to have GCK-MODY, whereas another was shown to have GCK-PNDM. A diagnosis of type 1 diabetes was confirmed in three relatives. This family exemplifies the importance of careful phenotyping and systematic evaluation of relatives after discovering monogenic diabetes in an individual.

    View details for DOI 10.1210/jc.2013-3641

    View details for Web of Science ID 000342340500019

    View details for PubMedID 24606082

    View details for PubMedCentralID PMC4186945

  • Analysis of the co-operative interaction between the allosterically regulated proteins GK and GKRP using tryptophan fluorescence BIOCHEMICAL JOURNAL Zelent, B., Raimondo, A., Barrettt, A., Buettger, C. W., Chen, P., Gloyn, A. L., Matschinsky, F. M. 2014; 459: 551–64

    Abstract

    Hepatic glucose phosphorylation by GK (glucokinase) is regulated by GKRP (GK regulatory protein). GKRP forms a cytosolic complex with GK followed by nuclear import and storage, leading to inhibition of GK activity. This process is initiated by low glucose, but reversed nutritionally by high glucose and fructose or pharmacologically by GKAs (GK activators) and GKRPIs (GKRP inhibitors). To study the regulation of this process by glucose, fructose-phosphate esters and a GKA, we measured the TF (tryptophan fluorescence) of human WT (wild-type) and GKRP-P446L (a mutation associated with high serum triacylglycerol) in the presence of non-fluorescent GK with its tryptophan residues mutated. Titration of GKRP-WT by GK resulted in a sigmoidal increase in TF, suggesting co-operative PPIs (protein-protein interactions) perhaps due to the hysteretic nature of GK. The affinity of GK for GKRP was decreased and binding co-operativity increased by glucose, fructose 1-phosphate and GKA, reflecting disruption of the GK-GKRP complex. Similar studies with GKRP-P446L showed significantly different results compared with GKRP-WT, suggesting impairment of complex formation and nuclear storage. The results of the present TF-based biophysical analysis of PPIs between GK and GKRP suggest that hepatic glucose metabolism is regulated by a metabolite-sensitive drug-responsive co-operative molecular switch, involving complex formation between these two allosterically regulated proteins.

    View details for DOI 10.1042/BJ20131363

    View details for Web of Science ID 000335125600011

    View details for PubMedID 24568320

    View details for PubMedCentralID PMC4109836

  • A Panel of Diverse Assays to Interrogate the Interaction between Glucokinase and Glucokinase Regulatory Protein, Two Vital Proteins in Human Disease PLOS ONE Rees, M. G., Davis, M. I., Shen, M., Titus, S., Raimondo, A., Barrett, A., Gloyn, A. L., Collins, F. S., Simeonov, A. 2014; 9 (2): e89335

    Abstract

    Recent genetic and clinical evidence has implicated glucokinase regulatory protein (GKRP) in the pathogenesis of type 2 diabetes and related traits. The primary role of GKRP is to bind and inhibit hepatic glucokinase (GCK), a critically important protein in human health and disease that exerts a significant degree of control over glucose metabolism. As activation of GCK has been associated with improved glucose tolerance, perturbation of the GCK-GKRP interaction represents a potential therapeutic target for pharmacological modulation. Recent structural and kinetic advances are beginning to provide insight into the interaction of these two proteins. However, tools to comprehensively assess the GCK-GKRP interaction, particularly in the context of small molecules, would be a valuable resource. We therefore developed three robust and miniaturized assays for assessing the interaction between recombinant human GCK and GKRP: an HTRF assay, a diaphorase-coupled assay, and a luciferase-coupled assay. The assays are complementary, featuring distinct mechanisms of detection (luminescence, fluorescence, FRET). Two assays rely on GCK enzyme activity modulation by GKRP while the FRET-based assay measures the GCK-GKRP protein-protein interaction independent of GCK enzymatic substrates and activity. All three assays are scalable to low volumes in 1536-well plate format, with robust Z' factors (>0.7). Finally, as GKRP sequesters GCK in the hepatocyte nucleus at low glucose concentrations, we explored cellular models of GCK localization and translocation. Previous findings from freshly isolated rat hepatocytes were confirmed in cryopreserved rat hepatocytes, and we further extended this study to cryopreserved human hepatocytes. Consistent with previous reports, there were several key differences between the rat and human systems, with our results suggesting that human hepatocytes can be used to interrogate GCK translocation in response to small molecules. The assay panel developed here should help direct future investigation of the GCK-GKRP interaction in these or other physiologically relevant human systems.

    View details for DOI 10.1371/journal.pone.0089335

    View details for Web of Science ID 000331711900126

    View details for PubMedID 24586696

    View details for PubMedCentralID PMC3929664

  • Pancreatic islet enhancer clusters enriched in type 2 diabetes risk-associated variants NATURE GENETICS Pasquali, L., Gaulton, K. J., Rodriguez-Segui, S. A., Mularoni, L., Miguel-Escalada, I., Akerman, I., Tena, J. J., Moran, I., Gomez-Marin, C., van de Bunt, M., Ponsa-Cobas, J., Castro, N., Nammo, T., Cebola, I., Garcia-Hurtado, J., Angel Maestro, M., Pattou, F., Piemonti, L., Berney, T., Gloyn, A. L., Ravassard, P., Luis Gomez-Skarmeta, J., Mueller, F., McCarthy, M. I., Ferrer, J. 2014; 46 (2): 136-+

    Abstract

    Type 2 diabetes affects over 300 million people, causing severe complications and premature death, yet the underlying molecular mechanisms are largely unknown. Pancreatic islet dysfunction is central in type 2 diabetes pathogenesis, and understanding islet genome regulation could therefore provide valuable mechanistic insights. We have now mapped and examined the function of human islet cis-regulatory networks. We identify genomic sequences that are targeted by islet transcription factors to drive islet-specific gene activity and show that most such sequences reside in clusters of enhancers that form physical three-dimensional chromatin domains. We find that sequence variants associated with type 2 diabetes and fasting glycemia are enriched in these clustered islet enhancers and identify trait-associated variants that disrupt DNA binding and islet enhancer activity. Our studies illustrate how islet transcription factors interact functionally with the epigenome and provide systematic evidence that the dysregulation of islet enhancers is relevant to the mechanisms underlying type 2 diabetes.

    View details for DOI 10.1038/ng.2870

    View details for Web of Science ID 000331208300009

    View details for PubMedID 24413736

    View details for PubMedCentralID PMC3935450

  • Argonaute2 Mediates Compensatory Expansion of the Pancreatic beta Cell CELL METABOLISM Tattikota, S. G., Rathjen, T., McAnulty, S. J., Wessels, H., Akerman, I., van de Bunt, M., Hausser, J., Esguerra, J. S., Musahl, A., Pandey, A. K., You, X., Chen, W., Herrera, P. L., Johnson, P. R., O'Carroll, D., Eliasson, L., Zavolan, M., Gloyn, A. L., Ferrer, J., Shalom-Feuerstein, R., Aberdam, D., Poy, M. N. 2014; 19 (1): 122–34

    Abstract

    Pancreatic β cells adapt to compensate for increased metabolic demand during insulin resistance. Although the microRNA pathway has an essential role in β cell proliferation, the extent of its contribution is unclear. Here, we report that miR-184 is silenced in the pancreatic islets of insulin-resistant mouse models and type 2 diabetic human subjects. Reduction of miR-184 promotes the expression of its target Argonaute2 (Ago2), a component of the microRNA-induced silencing complex. Moreover, restoration of miR-184 in leptin-deficient ob/ob mice decreased Ago2 and prevented compensatory β cell expansion. Loss of Ago2 during insulin resistance blocked β cell growth and relieved the regulation of miR-375-targeted genes, including the growth suppressor Cadm1. Lastly, administration of a ketogenic diet to ob/ob mice rescued insulin sensitivity and miR-184 expression and restored Ago2 and β cell mass. This study identifies the targeting of Ago2 by miR-184 as an essential component of the compensatory response to regulate proliferation according to insulin sensitivity.

    View details for DOI 10.1016/j.cmet.2013.11.015

    View details for Web of Science ID 000329431200014

    View details for PubMedID 24361012

    View details for PubMedCentralID PMC3945818

  • Phenotypic severity of homozygous GCK mutations causing neonatal or childhood-onset diabetes is primarily mediated through effects on protein stability. Human molecular genetics Raimondo, A. n., Chakera, A. J., Thomsen, S. K., Colclough, K. n., Barrett, A. n., De Franco, E. n., Chatelas, A. n., Demirbilek, H. n., Akcay, T. n., Alawneh, H. n., Flanagan, S. E., Van De Bunt, M. n., Hattersley, A. T., Gloyn, A. L., Ellard, S. n. 2014; 23 (24): 6432–40

    Abstract

    Mutations in glucokinase (GCK) cause a spectrum of glycemic disorders. Heterozygous loss-of-function mutations cause mild fasting hyperglycemia irrespective of mutation severity due to compensation from the unaffected allele. Conversely, homozygous loss-of-function mutations cause permanent neonatal diabetes requiring lifelong insulin treatment. This study aimed to determine the relationship between in vitro mutation severity and clinical phenotype in a large international case series of patients with homozygous GCK mutations. Clinical characteristics for 30 patients with diabetes due to homozygous GCK mutations (19 unique mutations, including 16 missense) were compiled and assigned a clinical severity grade (CSG) based on birth weight and age at diagnosis. The majority (28 of 30) of subjects were diagnosed before 9 months, with the remaining two at 9 and 15 years. These are the first two cases of a homozygous GCK mutation diagnosed outside infancy. Recombinant mutant GCK proteins were analyzed for kinetic and thermostability characteristics and assigned a relative activity index (RAI) or relative stability index (RSI) value. Six of 16 missense mutations exhibited severe kinetic defects (RAI ≤ 0.01). There was no correlation between CSG and RAI (r(2) = 0.05, P = 0.39), indicating that kinetics alone did not explain the phenotype. Eighty percent of the remaining mutations showed reduced thermostability, the exceptions being the two later-onset mutations which exhibited increased thermostability. Comparison of CSG with RSI detected a highly significant correlation (r(2) = 0.74, P = 0.002). We report the largest case series of homozygous GCK mutations to date and demonstrate that they can cause childhood-onset diabetes, with protein instability being the major determinant of mutation severity.

    View details for DOI 10.1093/hmg/ddu360

    View details for PubMedID 25015100

    View details for PubMedCentralID PMC4240195

  • Genetics in Diabetes Type 2 Diabetes and Related Traits Preface GENETICS IN DIABETES: TYPE 2 DIABETES AND RELATED TRAITS Gloyn, A. L., McCarthy, M. I., Gloyn, A. L., McCarthy, M. I. 2014; 23: VII
  • Translating Genetic Association Signals for Diabetes and Metabolic Traits into Molecular Mechanisms for Disease GENETICS IN DIABETES: TYPE 2 DIABETES AND RELATED TRAITS Rees, M. G., Gloyn, A. L., Gloyn, A. L., McCarthy, M. I. 2014; 23: 133–45

    View details for DOI 10.1159/000362473

    View details for Web of Science ID 000387711300012

  • Translating Advances in Our Understanding of the Genetics of Diabetes into the Clinic GENETICS IN DIABETES: TYPE 2 DIABETES AND RELATED TRAITS Gardner, D. S., Owen, K. R., Gloyn, A. L., Gloyn, A. L., McCarthy, M. I. 2014; 23: 173–86

    View details for DOI 10.1159/000362476

    View details for Web of Science ID 000387711300015

  • Inheritance of rare functional GCKR variants and their contribution to triglyceride levels in families. Human molecular genetics Rees, M. G., Raimondo, A. n., Wang, J. n., Ban, M. R., Davis, M. I., Barrett, A. n., Ranft, J. n., Jagdhuhn, D. n., Waterstradt, R. n., Baltrusch, S. n., Simeonov, A. n., Collins, F. S., Hegele, R. A., Gloyn, A. L. 2014; 23 (20): 5570–78

    Abstract

    Significant resources have been invested in sequencing studies to investigate the role of rare variants in complex disease etiology. However, the diagnostic interpretation of individual rare variants remains a major challenge, and may require accurate variant functional classification and the collection of large numbers of variant carriers. Utilizing sequence data from 458 individuals with hypertriglyceridemia and 333 controls with normal plasma triglyceride levels, we investigated these issues using GCKR, encoding glucokinase regulatory protein. Eighteen rare non-synonymous GCKR variants identified in these 791 individuals were comprehensively characterized by a range of biochemical and cell biological assays, including a novel high-throughput-screening-based approach capable of measuring all variant proteins simultaneously. Functionally deleterious variants were collectively associated with hypertriglyceridemia, but a range of in silico prediction algorithms showed little consistency between algorithms and poor agreement with functional data. We extended our study by obtaining sequence data on family members; however, functional variants did not co-segregate with triglyceride levels. Therefore, despite evidence for their collective functional and clinical relevance, our results emphasize the low predictive value of rare GCKR variants in individuals and the complex heritability of lipid traits.

    View details for DOI 10.1093/hmg/ddu269

    View details for PubMedID 24879641

    View details for PubMedCentralID PMC4168830

  • Role of K-ATP Channels in Glucose-Regulated Glucagon Secretion and Impaired Counterregulation in Type 2 Diabetes CELL METABOLISM Zhang, Q., Ramracheya, R., Lahmann, C., Tarasov, A., Bengtsson, M., Braha, O., Braun, M., Brereton, M., Collins, S., Galvanovskis, J., Gonzalez, A., Groschner, L. N., Rorsman, N. G., Salehi, A., Travers, M. E., Walker, J. N., Gloyn, A. L., Gribble, F., Johnson, P. V., Reimann, F., Ashcroft, F. M., Rorsman, P. 2013; 18 (6): 871–82

    Abstract

    Glucagon, secreted by pancreatic islet α cells, is the principal hyperglycemic hormone. In diabetes, glucagon secretion is not suppressed at high glucose, exacerbating the consequences of insufficient insulin secretion, and is inadequate at low glucose, potentially leading to fatal hypoglycemia. The causal mechanisms remain unknown. Here we show that α cell KATP-channel activity is very low under hypoglycemic conditions and that hyperglycemia, via elevated intracellular ATP/ADP, leads to complete inhibition. This produces membrane depolarization and voltage-dependent inactivation of the Na(+) channels involved in action potential firing that, via reduced action potential height and Ca(2+) entry, suppresses glucagon secretion. Maneuvers that increase KATP channel activity, such as metabolic inhibition, mimic the glucagon secretory defects associated with diabetes. Low concentrations of the KATP channel blocker tolbutamide partially restore glucose-regulated glucagon secretion in islets from type 2 diabetic organ donors. These data suggest that impaired metabolic control of the KATP channels underlies the defective glucose regulation of glucagon secretion in type 2 diabetes.

    View details for DOI 10.1016/j.cmet.2013.10.014

    View details for Web of Science ID 000327940800012

    View details for PubMedID 24315372

    View details for PubMedCentralID PMC3851686

  • Bridging the Gap Between Genetic Associations and Molecular Mechanisms for Type 2 Diabetes CURRENT DIABETES REPORTS Ng, H., Gloyn, A. L. 2013; 13 (6): 778–85

    Abstract

    Type 2 diabetes is a global pandemic for which there is currently no disease-modifying treatment. New and targeted therapeutics are greatly needed, but progress in identifying novel targets for therapeutic intervention is severely hampered by poor understanding of disease pathogenesis. Over the past 6 years, the success of genome-wide association studies has led to an unprecedented increase in the number of loci robustly associating with type 2 diabetes risk. Each of these signals offers the opportunity to uncover biological insights into disease pathogenesis, which, if harnessed effectively, hold the promise to deliver new pathways for therapeutic intervention, strategies for patient stratification, and potentially, biomarkers for identifying those at greatest risk of developing diabetes. We review the progress that has been made and the approaches being adopted and discuss the inherent challenges in moving from association signals, which largely map to poorly annotated sequence, to transcripts, mechanisms, and disease biology.

    View details for DOI 10.1007/s11892-013-0429-1

    View details for Web of Science ID 000326188500003

    View details for PubMedID 24127137

  • Genome-Wide Association Study Identifies a Novel Locus Contributing to Type 2 Diabetes Susceptibility in Sikhs of Punjabi Origin From India DIABETES Saxena, R., Saleheen, D., Been, L. F., Garavito, M. L., Braun, T., Bjonnes, A., Young, R., Ho, W., Rasheed, A., Frossard, P., Sim, X., Hassanali, N., Radha, V., Chidambaram, M., Liju, S., Rees, S. D., Ng, D., Wong, T., Yamauchi, T., Hara, K., Tanaka, Y., Hirose, H., McCarthy, M. I., Morris, A. P., Basit, A., Barnett, A. H., Katulanda, P., Matthews, D., Mohan, V., Wander, G. S., Singh, J., Mehra, N. K., Ralhan, S., Kamboh, M., Mulvihill, J. J., Maegawa, H., Tobe, K., Maeda, S., Cho, Y. S., Tai, E., Kelly, M., Chambers, J. C., Kooner, J. S., Kadowaki, T., Deloukas, P., Rader, D. J., Danesh, J., Sanghera, D. K., DIAGRAM, MuTHER, AGEN 2013; 62 (5): 1746–55

    Abstract

    We performed a genome-wide association study (GWAS) and a multistage meta-analysis of type 2 diabetes (T2D) in Punjabi Sikhs from India. Our discovery GWAS in 1,616 individuals (842 case subjects) was followed by in silico replication of the top 513 independent single nucleotide polymorphisms (SNPs) (P < 10⁻³) in Punjabi Sikhs (n = 2,819; 801 case subjects). We further replicated 66 SNPs (P < 10⁻⁴) through genotyping in a Punjabi Sikh sample (n = 2,894; 1,711 case subjects). On combined meta-analysis in Sikh populations (n = 7,329; 3,354 case subjects), we identified a novel locus in association with T2D at 13q12 represented by a directly genotyped intronic SNP (rs9552911, P = 1.82 × 10⁻⁸) in the SGCG gene. Next, we undertook in silico replication (stage 2b) of the top 513 signals (P < 10⁻³) in 29,157 non-Sikh South Asians (10,971 case subjects) and de novo genotyping of up to 31 top signals (P < 10⁻⁴) in 10,817 South Asians (5,157 case subjects) (stage 3b). In combined South Asian meta-analysis, we observed six suggestive associations (P < 10⁻⁵ to < 10⁻⁷), including SNPs at HMG1L1/CTCFL, PLXNA4, SCAP, and chr5p11. Further evaluation of 31 top SNPs in 33,707 East Asians (16,746 case subjects) (stage 3c) and 47,117 Europeans (8,130 case subjects) (stage 3d), and joint meta-analysis of 128,127 individuals (44,358 case subjects) from 27 multiethnic studies, did not reveal any additional loci nor was there any evidence of replication for the new variant. Our findings provide new evidence on the presence of a population-specific signal in relation to T2D, which may provide additional insights into T2D pathogenesis.

    View details for DOI 10.2337/db12-1077

    View details for Web of Science ID 000318128700054

    View details for PubMedID 23300278

    View details for PubMedCentralID PMC3636649

  • Mutations in HNF1A Result in Marked Alterations of Plasma Glycan Profile DIABETES Thanabalasingham, G., Huffman, J. E., Kattla, J. J., Novokmet, M., Rudan, I., Gloyn, A. L., Hayward, C., Adamczyk, B., Reynolds, R. M., Muzinic, A., Hassanali, N., Pucic, M., Bennett, A. J., Essafi, A., Polasek, O., Mughal, S. A., Redzic, I., Primorac, D., Zgaga, L., Kokic, I., Hansen, T., Gasperikova, D., Tjora, E., Strachan, M. J., Nielsen, T., Stanik, J., Klimes, I., Pedersen, O. B., Njolstad, P. R., Wild, S. H., Gyllensten, U., Gornik, O., Wilson, J. F., Hastie, N. D., Campbell, H., McCarthy, M. I., Rudd, P. M., Owen, K. R., Lauc, G., Wright, A. F. 2013; 62 (4): 1329–37

    Abstract

    A recent genome-wide association study identified hepatocyte nuclear factor 1-α (HNF1A) as a key regulator of fucosylation. We hypothesized that loss-of-function HNF1A mutations causal for maturity-onset diabetes of the young (MODY) would display altered fucosylation of N-linked glycans on plasma proteins and that glycan biomarkers could improve the efficiency of a diagnosis of HNF1A-MODY. In a pilot comparison of 33 subjects with HNF1A-MODY and 41 subjects with type 2 diabetes, 15 of 29 glycan measurements differed between the two groups. The DG9-glycan index, which is the ratio of fucosylated to nonfucosylated triantennary glycans, provided optimum discrimination in the pilot study and was examined further among additional subjects with HNF1A-MODY (n = 188), glucokinase (GCK)-MODY (n = 118), hepatocyte nuclear factor 4-α (HNF4A)-MODY (n = 40), type 1 diabetes (n = 98), type 2 diabetes (n = 167), and nondiabetic controls (n = 98). The DG9-glycan index was markedly lower in HNF1A-MODY than in controls or other diabetes subtypes, offered good discrimination between HNF1A-MODY and both type 1 and type 2 diabetes (C statistic ≥ 0.90), and enabled us to detect three previously undetected HNF1A mutations in patients with diabetes. In conclusion, glycan profiles are altered substantially in HNF1A-MODY, and the DG9-glycan index has potential clinical value as a diagnostic biomarker of HNF1A dysfunction.

    View details for DOI 10.2337/db12-0880

    View details for Web of Science ID 000316526000043

    View details for PubMedID 23274891

    View details for PubMedCentralID PMC3609552

  • Genome-Wide Association Study for Type 2 Diabetes in Indians Identifies a New Susceptibility Locus at 2q21 DIABETES Tabassum, R., Chauhan, G., Dwivedi, O., Mahajan, A., Jaiswal, A., Kaur, I., Bandesh, K., Singh, T., Mathai, B., Pandey, Y., Chidambaram, M., Sharma, A., Chavali, S., Sengupta, S., Ramakrishnan, L., Venkatesh, P., Aggarwal, S. K., Ghosh, S., Prabhakaran, D., Srinath, R. K., Saxena, M., Banerjee, M., Mathur, S., Bhansali, A., Shah, V. N., Madhu, S., Marwaha, R. K., Basu, A., Scaria, V., McCarthy, M. I., Venkatesan, R., Mohan, V., Tandon, N., Bharadwaj, D., DIAGRAM, INDICO 2013; 62 (3): 977–86

    Abstract

    Indians undergoing socioeconomic and lifestyle transitions will be maximally affected by epidemic of type 2 diabetes (T2D). We conducted a two-stage genome-wide association study of T2D in 12,535 Indians, a less explored but high-risk group. We identified a new type 2 diabetes-associated locus at 2q21, with the lead signal being rs6723108 (odds ratio 1.31; P = 3.32 × 10⁻⁹). Imputation analysis refined the signal to rs998451 (odds ratio 1.56; P = 6.3 × 10⁻¹²) within TMEM163 that encodes a probable vesicular transporter in nerve terminals. TMEM163 variants also showed association with decreased fasting plasma insulin and homeostatic model assessment of insulin resistance, indicating a plausible effect through impaired insulin secretion. The 2q21 region also harbors RAB3GAP1 and ACMSD; those are involved in neurologic disorders. Forty-nine of 56 previously reported signals showed consistency in direction with similar effect sizes in Indians and previous studies, and 25 of them were also associated (P < 0.05). Known loci and the newly identified 2q21 locus altogether explained 7.65% variance in the risk of T2D in Indians. Our study suggests that common susceptibility variants for T2D are largely the same across populations, but also reveals a population-specific locus and provides further insights into genetic architecture and etiology of T2D.

    View details for DOI 10.2337/db12-0406

    View details for Web of Science ID 000315556400041

    View details for PubMedID 23209189

    View details for PubMedCentralID PMC3581193

  • TCF7L2 and Diabetes: A Tale of Two Tissues, and of Two Species CELL METABOLISM McCarthy, M. I., Rorsman, P., Gloyn, A. L. 2013; 17 (2): 157–59

    Abstract

    Human genetics is revealing ever more variants that influence propensity to common diseases, but progress in translating these discoveries into the biological mechanisms responsible for predisposition continues to lag behind. A recent paper in Cell (Boj et al., 2012) using rodent models to examine how diabetes-associated variants near TCF7L2 perturb metabolic regulation provides surprising results.

    View details for DOI 10.1016/j.cmet.2013.01.011

    View details for Web of Science ID 000326265000003

    View details for PubMedID 23395164

  • Apolipoprotein M can discriminate HNF1A-MODY from Type 1 diabetes DIABETIC MEDICINE Mughal, S. A., Park, R., Nowak, N., Gloyn, A. L., Karpe, F., Matile, H., Malecki, M. T., McCarthy, M. I., Stoffel, M., Owen, K. R. 2013; 30 (2): 246–50

    Abstract

    Missed diagnosis of maturity-onset diabetes of the young (MODY) has led to an interest in biomarkers that enable efficient prioritization of patients for definitive molecular testing. Apolipoprotein M (apoM) was suggested as a biomarker for hepatocyte nuclear factor 1 alpha (HNF1A)-MODY because of its reduced expression in Hnf1a(-/-) mice. However, subsequent human studies examining apoM as a biomarker have yielded conflicting results. We aimed to evaluate apoM as a biomarker for HNF1A-MODY using a highly specific and sensitive ELISA.ApoM concentration was measured in subjects with HNF1A-MODY (n = 69), Type 1 diabetes (n = 50), Type 2 diabetes (n = 120) and healthy control subjects (n = 100). The discriminative accuracy of apoM and of the apoM/HDL ratio for diabetes aetiology was evaluated.Mean (standard deviation) serum apoM concentration (μmol/l) was significantly lower for subjects with HNF1A-MODY [0.86 (0.29)], than for those with Type 1 diabetes [1.37 (0.26), P = 3.1 × 10(-18) ) and control subjects [1.34 (0.22), P = 7.2 × 10(-19) ). There was no significant difference in apoM concentration between subjects with HNF1A-MODY and Type 2 diabetes [0.89 (0.28), P = 0.13]. The C-statistic measure of discriminative accuracy for apoM was 0.91 for HNF1A-MODY vs. Type 1 diabetes, indicating high discriminative accuracy. The apoM/HDL ratio was significantly lower in HNF1A-MODY than other study groups. However, this ratio did not perform well in discriminating HNF1A-MODY from either Type 1 diabetes (C-statistic = 0.79) or Type 2 diabetes (C-statistic = 0.68).We confirm an earlier report that serum apoM levels are lower in HNF1A-MODY than in controls. Serum apoM provides good discrimination between HNF1A-MODY and Type 1 diabetes and warrants further investigation for clinical utility in diabetes diagnostics.

    View details for DOI 10.1111/dme.12066

    View details for Web of Science ID 000313876500021

    View details for PubMedID 23157689

    View details for PubMedCentralID PMC4193536

  • The miRNA Profile of Human Pancreatic Islets and Beta-Cells and Relationship to Type 2 Diabetes Pathogenesis PLOS ONE van de Bunt, M., Gaulton, K. J., Parts, L., Moran, I., Johnson, P. R., Lindgren, C. M., Ferrer, J., Gloyn, A. L., McCarthy, M. I. 2013; 8 (1): e55272

    Abstract

    Recent advances in the understanding of the genetics of type 2 diabetes (T2D) susceptibility have focused attention on the regulation of transcriptional activity within the pancreatic beta-cell. MicroRNAs (miRNAs) represent an important component of regulatory control, and have proven roles in the development of human disease and control of glucose homeostasis. We set out to establish the miRNA profile of human pancreatic islets and of enriched beta-cell populations, and to explore their potential involvement in T2D susceptibility. We used Illumina small RNA sequencing to profile the miRNA fraction in three preparations each of primary human islets and of enriched beta-cells generated by fluorescence-activated cell sorting. In total, 366 miRNAs were found to be expressed (i.e. >100 cumulative reads) in islets and 346 in beta-cells; of the total of 384 unique miRNAs, 328 were shared. A comparison of the islet-cell miRNA profile with those of 15 other human tissues identified 40 miRNAs predominantly expressed (i.e. >50% of all reads seen across the tissues) in islets. Several highly-expressed islet miRNAs, such as miR-375, have established roles in the regulation of islet function, but others (e.g. miR-27b-3p, miR-192-5p) have not previously been described in the context of islet biology. As a first step towards exploring the role of islet-expressed miRNAs and their predicted mRNA targets in T2D pathogenesis, we looked at published T2D association signals across these sites. We found evidence that predicted mRNA targets of islet-expressed miRNAs were globally enriched for signals of T2D association (p-values <0.01, q-values <0.1). At six loci with genome-wide evidence for T2D association (AP3S2, KCNK16, NOTCH2, SCL30A8, VPS26A, and WFS1) predicted mRNA target sites for islet-expressed miRNAs overlapped potentially causal variants. In conclusion, we have described the miRNA profile of human islets and beta-cells and provide evidence linking islet miRNAs to T2D pathogenesis.

    View details for DOI 10.1371/journal.pone.0055272

    View details for Web of Science ID 000315210400086

    View details for PubMedID 23372846

    View details for PubMedCentralID PMC3555946

  • Insights into the molecular mechanism for type 2 diabetes susceptibility at the KCNQ1 locus from temporal changes in imprinting status in human islets. Diabetes Travers, M. E., Mackay, D. J., Dekker Nitert, M. n., Morris, A. P., Lindgren, C. M., Berry, A. n., Johnson, P. R., Hanley, N. n., Groop, L. C., McCarthy, M. I., Gloyn, A. L. 2013; 62 (3): 987–92

    Abstract

    The molecular basis of type 2 diabetes predisposition at most established susceptibility loci remains poorly understood. KCNQ1 maps within the 11p15.5 imprinted domain, a region with an established role in congenital growth phenotypes. Variants intronic to KCNQ1 influence diabetes susceptibility when maternally inherited. By use of quantitative PCR and pyrosequencing of human adult islet and fetal pancreas samples, we investigated the imprinting status of regional transcripts and aimed to determine whether type 2 diabetes risk alleles influence regional DNA methylation and gene expression. The results demonstrate that gene expression patterns differ by developmental stage. CDKN1C showed monoallelic expression in both adult and fetal tissue, whereas PHLDA2, SLC22A18, and SLC22A18AS were biallelically expressed in both tissues. Temporal changes in imprinting were observed for KCNQ1 and KCNQ1OT1, with monoallelic expression in fetal tissues and biallelic expression in adult samples. Genotype at the type 2 diabetes risk variant rs2237895 influenced methylation levels of regulatory sequence in fetal pancreas but without demonstrable effects on gene expression. We demonstrate that CDKN1C, KCNQ1, and KCNQ1OT1 are most likely to mediate diabetes susceptibility at the KCNQ1 locus and identify temporal differences in imprinting status and methylation effects, suggesting that diabetes risk effects may be mediated in early development.

    View details for DOI 10.2337/db12-0819

    View details for PubMedID 23139357

    View details for PubMedCentralID PMC3581222

  • Small molecular glucokinase activators: has another new anti-diabetic therapeutic lost favour? BRITISH JOURNAL OF PHARMACOLOGY Rees, M. G., Gloyn, A. L. 2013; 168 (2): 335–38

    Abstract

    Glucokinase activators (GKAs) represent one of the leading hopes for the next generation of type 2 diabetes (T2D) therapeutics, showing efficacy in reducing blood glucose and HbA1c levels in animal models of T2D and short-term human trials. While the hypoglycaemic risks of GCK activation in pancreatic beta-cells have long been appreciated, the hepatic effects of GKAs have generally been perceived to be without significant side effect. In this issue of the British Journal of Pharmacology, De Ceuninck et al. report that acute and chronic GKA treatment of normoglycaemic and hyperglycaemic rodent models results in significant accumulation of triglycerides in the liver. This suggests GKA-mediated activation of hepatic glucose uptake and suppression of endogenous glucose production may come at a significant cost; namely, the development of hepatic steatosis. This raises important questions regarding the safety of GKAs and emphasizes that both plasma and hepatic lipid profiles should be carefully monitored in on-going and future studies of these molecules.This article is a commentary on De Ceuninck et al., pp. 339-353 of this issue. To view this paper visit http://dx.doi.org/10.1111/j.1476-5381.2012.02184.x.

    View details for DOI 10.1111/j.1476-5381.2012.02201.x

    View details for Web of Science ID 000313751200006

    View details for PubMedID 22946641

    View details for PubMedCentralID PMC3572560

  • SSTR2 is the functionally dominant somatostatin receptor in human pancreatic beta- and alpha-cells AMERICAN JOURNAL OF PHYSIOLOGY-ENDOCRINOLOGY AND METABOLISM Kailey, B., van de Bunt, M., Cheley, S., Johnson, P. R., MacDonald, P. E., Gloyn, A. L., Rorsman, P., Braun, M. 2012; 303 (9): E1107–E1116

    Abstract

    Somatostatin-14 (SST) inhibits insulin and glucagon secretion by activating G protein-coupled somatostatin receptors (SSTRs), of which five isoforms exist (SSTR1-5). In mice, the effects on pancreatic β-cells are mediated by SSTR5, whereas α-cells express SSTR2. In both cell types, SSTR activation results in membrane hyperpolarization and suppression of exocytosis. Here, we examined the mechanisms by which SST inhibits secretion from human β- and α-cells and the SSTR isoforms mediating these effects. Quantitative PCR revealed high expression of SSTR2, with lower levels of SSTR1, SSTR3, and SSTR5, in human islets. Immunohistochemistry showed expression of SSTR2 in both β- and α-cells. SST application hyperpolarized human β-cells and inhibited action potential firing. The membrane hyperpolarization was unaffected by tolbutamide but antagonized by tertiapin-Q, a blocker of G protein-gated inwardly rectifying K⁺ channels (GIRK). The effect of SST was mimicked by an SSTR2-selective agonist, whereas a SSTR5 agonist was marginally effective. SST strongly (>70%) reduced depolarization-evoked exocytosis in both β- and α-cells. A slightly weaker inhibition was observed in both cell types after SSTR2 activation. SSTR3- and SSTR1-selective agonists moderately reduced the exocytotic responses in β- and α-cells, respectively, whereas SSTR4- and SSTR5-specific agonists were ineffective. SST also reduced voltage-gated P/Q-type Ca²⁺ currents in β-cells, but normalization of Ca²⁺ influx to control levels by prolonged depolarizations only partially restored exocytosis. We conclude that SST inhibits secretion from both human β- and α-cells by activating GIRK and suppressing electrical activity, reducing P/Q-type Ca²⁺ currents, and directly inhibiting exocytosis. These effects are mediated predominantly by SSTR2 in both cell types.

    View details for DOI 10.1152/ajpendo.00207.2012

    View details for Web of Science ID 000310647800003

    View details for PubMedID 22932785

    View details for PubMedCentralID PMC3492856

  • Human beta Cell Transcriptome Analysis Uncovers IncRNAs That Are Tissue-Specific, Dynamically Regulated, and Abnormally Expressed in Type 2 Diabetes CELL METABOLISM Moran, I., Akerman, I., van de Bunt, M., Xie, R., Benazra, M., Nammo, T., Arnes, L., Nakic, N., Garcia-Hurtado, J., Rodriguez-Segui, S., Pasquali, L., Sauty-Colace, C., Beucher, A., Scharfmann, R., van Arensbergen, J., Johnson, P. R., Berry, A., Lee, C., Harkins, T., Gmyr, V., Pattou, F., Kerr-Conte, J., Piemonti, L., Berney, T., Hanley, N., Gloyn, A. L., Sussel, L., Langman, L., Brayman, K. L., Sander, M., McCarthy, M. I., Ravassard, P., Ferrer, J. 2012; 16 (4): 435–48

    Abstract

    A significant portion of the genome is transcribed as long noncoding RNAs (lncRNAs), several of which are known to control gene expression. The repertoire and regulation of lncRNAs in disease-relevant tissues, however, has not been systematically explored. We report a comprehensive strand-specific transcriptome map of human pancreatic islets and β cells, and uncover >1100 intergenic and antisense islet-cell lncRNA genes. We find islet lncRNAs that are dynamically regulated and show that they are an integral component of the β cell differentiation and maturation program. We sequenced the mouse islet transcriptome and identify lncRNA orthologs that are regulated like their human counterparts. Depletion of HI-LNC25, a β cell-specific lncRNA, downregulated GLIS3 mRNA, thus exemplifying a gene regulatory function of islet lncRNAs. Finally, selected islet lncRNAs were dysregulated in type 2 diabetes or mapped to genetic loci underlying diabetes susceptibility. These findings reveal a new class of islet-cell genes relevant to β cell programming and diabetes pathophysiology.

    View details for DOI 10.1016/j.cmet.2012.08.010

    View details for Web of Science ID 000309948400007

    View details for PubMedID 23040067

    View details for PubMedCentralID PMC3475176

  • A tale of two glucose transporters: how GLUT2 re-emerged as a contender for glucose transport into the human beta cell DIABETOLOGIA van de Bunt, M., Gloyn, A. L. 2012; 55 (9): 2312–15

    Abstract

    Finding novel causes for monogenic forms of diabetes is important as, alongside the clinical implications of such a discovery, it can identify critical proteins and pathways required for normal beta cell function in humans. It is increasingly apparent that there are significant differences between rodent and human islets. One example that has generated interest is the relative importance of the glucose transporter GLUT2 in rodent and human beta cells. The central role of GLUT2 in rodent beta cells is well established, but a number of studies have suggested that other glucose transporters, namely GLUT1 and GLUT3, may play an important role in facilitating glucose transport into human beta cells. In this issue of Diabetologia Sansbury et al (DOI: 10.1007/s00125-012-2595-0 ) report homozygous loss of function mutations in SLC2A2, which encodes GLUT2, as a rare cause of neonatal diabetes. Evidence for a beta cell defect in these subjects comes from very low birthweights, lack of endogenous insulin secretion and a requirement for insulin therapy. Neonatal diabetes is not a consistent feature of SLC2A2 mutations. It is only found in a small percentage of cases (~4%) and the diabetes largely resolves before 18 months of age. This discovery is significant as it suggests that GLUT2 plays an important role in human beta cells, but the interplay and relative roles of other transporters differ from those in rodents. This finding should encourage efforts to delineate the precise role of GLUT2 in the human beta cell at different developmental time points and is a further reminder of critical differences between human and rodent islets.

    View details for DOI 10.1007/s00125-012-2612-3

    View details for Web of Science ID 000307301800003

    View details for PubMedID 22696037

  • Metabolic Profiling in Maturity-Onset Diabetes of the Young (MODY) and Young Onset Type 2 Diabetes Fails to Detect Robust Urinary Biomarkers PLOS ONE Gloyn, A. L., Faber, J. H., Malmodin, D., Thanabalasingham, G., Lam, F., Ueland, P., McCarthy, M. I., Owen, K. R., Baunsgaard, D. 2012; 7 (7): e40962

    Abstract

    It is important to identify patients with Maturity-onset diabetes of the young (MODY) as a molecular diagnosis determines both treatment and prognosis. Genetic testing is currently expensive and many patients are therefore not assessed and are misclassified as having either type 1 or type 2 diabetes. Biomarkers could facilitate the prioritisation of patients for genetic testing. We hypothesised that patients with different underlying genetic aetiologies for their diabetes could have distinct metabolic profiles which may uncover novel biomarkers. The aim of this study was to perform metabolic profiling in urine from patients with MODY due to mutations in the genes encoding glucokinase (GCK) or hepatocyte nuclear factor 1 alpha (HNF1A), type 2 diabetes (T2D) and normoglycaemic control subjects. Urinary metabolic profiling by Nuclear Magnetic Resonance (NMR) and ultra performance liquid chromatography hyphenated to Q-TOF mass spectrometry (UPLC-MS) was performed in a Discovery set of subjects with HNF1A-MODY (n = 14), GCK-MODY (n = 17), T2D (n = 14) and normoglycaemic controls (n = 34). Data were used to build a valid partial least squares discriminate analysis (PLS-DA) model where HNF1A-MODY subjects could be separated from the other diabetes subtypes. No single metabolite contributed significantly to the separation of the patient groups. However, betaine, valine, glycine and glucose were elevated in the urine of HNF1A-MODY subjects compared to the other subgroups. Direct measurements of urinary amino acids and betaine in an extended dataset did not support differences between patients groups. Elevated urinary glucose in HNF1A-MODY is consistent with the previously reported low renal threshold for glucose in this genetic subtype. In conclusion, we report the first metabolic profiling study in monogenic diabetes and show that, despite the distinct biochemical pathways affected, there are unlikely to be robust urinary biomarkers which distinguish monogenic subtypes from T2D. Our results have implications for studies investigating metabolic profiles in complex traits including T2D.

    View details for DOI 10.1371/journal.pone.0040962

    View details for Web of Science ID 000306950900014

    View details for PubMedID 22859960

    View details for PubMedCentralID PMC3408469

  • Reduced Insulin Exocytosis in Human Pancreatic beta-Cells With Gene Variants Linked to Type 2 Diabetes DIABETES Rosengren, A. H., Braun, M., Mandi, T., Andersson, S. A., Travers, M. E., Shigeto, M., Zhang, E., Almgren, P., Ladenvall, C., Axelsson, A. S., Edlund, A., Pedersen, M., Jonsson, A., Ramracheya, R., Tang, Y., Walker, J. N., Barrett, A., Johnson, P. V., Lyssenko, V., McCarthy, M. I., Groop, L., Salehi, A., Gloyn, A. L., Renstrom, E., Rorsman, P., Eliasson, L. 2012; 61 (7): 1726–33

    Abstract

    The majority of genetic risk variants for type 2 diabetes (T2D) affect insulin secretion, but the mechanisms through which they influence pancreatic islet function remain largely unknown. We functionally characterized human islets to determine secretory, biophysical, and ultrastructural features in relation to genetic risk profiles in diabetic and nondiabetic donors. Islets from donors with T2D exhibited impaired insulin secretion, which was more pronounced in lean than obese diabetic donors. We assessed the impact of 14 disease susceptibility variants on measures of glucose sensing, exocytosis, and structure. Variants near TCF7L2 and ADRA2A were associated with reduced glucose-induced insulin secretion, whereas susceptibility variants near ADRA2A, KCNJ11, KCNQ1, and TCF7L2 were associated with reduced depolarization-evoked insulin exocytosis. KCNQ1, ADRA2A, KCNJ11, HHEX/IDE, and SLC2A2 variants affected granule docking. We combined our results to create a novel genetic risk score for β-cell dysfunction that includes aberrant granule docking, decreased Ca(2+) sensitivity of exocytosis, and reduced insulin release. Individuals with a high risk score displayed an impaired response to intravenous glucose and deteriorating insulin secretion over time. Our results underscore the importance of defects in β-cell exocytosis in T2D and demonstrate the potential of cellular phenotypic characterization in the elucidation of complex genetic disorders.

    View details for DOI 10.2337/db11-1516

    View details for Web of Science ID 000305543900013

    View details for PubMedID 22492527

    View details for PubMedCentralID PMC3379663

  • A genome-wide approach accounting for body mass index identifies genetic variants influencing fasting glycemic traits and insulin resistance NATURE GENETICS Manning, A. K., Hivert, M., Scott, R. A., Grimsby, J. L., Bouatia-Naji, N., Chen, H., Rybin, D., Liu, C., Bielak, L. F., Prokopenko, I., Amin, N., Barnes, D., Cadby, G., Hottenga, J., Ingelsson, E., Jackson, A. U., Johnson, T., Kanoni, S., Ladenvall, C., Lagou, V., Lahti, J., Lecoeur, C., Liu, Y., Martinez-Larrad, M. T., Montasser, M. E., Navarro, P., Perry, J. R., Rasmussen-Torvik, L. J., Salo, P., Sattar, N., Shungin, D., Strawbridge, R. J., Tanaka, T., van Duijn, C. M., An, P., de Andrade, M., Andrews, J. S., Aspelund, T., Atalay, M., Aulchenko, Y., Balkau, B., Bandinelli, S., Beckmann, J. S., Beilby, J. P., Bellis, C., Bergman, R. N., Blangero, J., Boban, M., Boehnke, M., Boerwinkle, E., Bonnycastle, L. L., Boomsma, D. I., Borecki, I. B., Boettcher, Y., Bouchard, C., Brunner, E., Budimir, D., Campbell, H., Carlson, O., Chines, P. S., Clarke, R., Collins, F. S., Corbaton-Anchuelo, A., Couper, D., de Faire, U., Dedoussis, G. V., Deloukas, P., Dimitriou, M., Egan, J. M., Eiriksdottir, G., Erdos, M. R., Eriksson, J. G., Eury, E., Ferrucci, L., Ford, I., Forouhi, N. G., Fox, C. S., Franzosi, M. G., Franks, P. W., Frayling, T. M., Froguel, P., Galan, P., de Geus, E., Gigante, B., Glazer, N. L., Goel, A., Groop, L., Gudnason, V., Hallmans, G., Hamsten, A., Hansson, O., Harris, T. B., Hayward, C., Heath, S., Hercberg, S., Hicks, A. A., Hingorani, A., Hofman, A., Hui, J., Hung, J., Jarvelin, M., Jhun, M. A., Johnson, P. C., Jukema, J. W., Jula, A., Kao, W. H., Kaprio, J., Kardia, S. L., Keinanen-Kiukaanniemi, S., Kivimaki, M., Kolcic, I., Kovacs, P., Kumari, M., Kuusisto, J., Kyvik, K. O., Laakso, M., Lakka, T., Lannfelt, L., Lathrop, G. M., Launer, L. J., Leander, K., Li, G., Lind, L., Lindstrom, J., Lobbens, S., Loos, R. J., Luan, J., Lyssenko, V., Magi, R., Magnusson, P. K., Marmot, M., Meneton, P., Mohlke, K. L., Mooser, V., Morken, M. A., Miljkovic, I., Narisu, N., O'Connell, J., Ong, K. K., Oostra, B. A., Palmer, L. J., Palotie, A., Pankow, J. S., Peden, J. F., Pedersen, N. L., Pehlic, M., Peltonen, L., Penninx, B., Pericic, M., Perola, M., Perusse, L., Peyser, P. A., Polasek, O., Pramstaller, P. P., Province, M. A., Raikkonen, K., Rauramaa, R., Rehnberg, E., Rice, K., Rotter, J. I., Rudan, I., Ruokonen, A., Saaristo, T., Sabater-Lleal, M., Salomaa, V., Savage, D. B., Saxena, R., Schwarz, P., Seedorf, U., Sennblad, B., Serrano-Rios, M., Shuldiner, A. R., Sijbrands, E. J., Siscovick, D. S., Smit, J. H., Small, K. S., Smith, N. L., Smith, A. V., Stancakova, A., Stirrups, K., Stumvoll, M., Sun, Y. V., Swift, A. J., Toenjes, A., Tuomilehto, J., Trompet, S., Uitterlinden, A. G., Uusitupa, M., Vikstrom, M., Vitart, V., Vohl, M., Voight, B. F., Vollenweider, P., Waeber, G., Waterworth, D. M., Watkins, H., Wheeler, E., Widen, E., Wild, S. H., Willems, S. M., Willemsen, G., Wilson, J. F., Witteman, J. C., Wright, A. F., Yaghootkar, H., Zelenika, D., Zemunik, T., Zgaga, L., Wareham, N. J., McCarthy, M. I., Barroso, I., Watanabe, R. M., Florez, J. C., Dupuis, J., Meigs, J. B., Langenberg, C. 2012; 44 (6): 659-U81

    Abstract

    Recent genome-wide association studies have described many loci implicated in type 2 diabetes (T2D) pathophysiology and β-cell dysfunction but have contributed little to the understanding of the genetic basis of insulin resistance. We hypothesized that genes implicated in insulin resistance pathways might be uncovered by accounting for differences in body mass index (BMI) and potential interactions between BMI and genetic variants. We applied a joint meta-analysis approach to test associations with fasting insulin and glucose on a genome-wide scale. We present six previously unknown loci associated with fasting insulin at P < 5 × 10(-8) in combined discovery and follow-up analyses of 52 studies comprising up to 96,496 non-diabetic individuals. Risk variants were associated with higher triglyceride and lower high-density lipoprotein (HDL) cholesterol levels, suggesting a role for these loci in insulin resistance pathways. The discovery of these loci will aid further characterization of the role of insulin resistance in T2D pathophysiology.

    View details for DOI 10.1038/ng.2274

    View details for Web of Science ID 000304551100012

    View details for PubMedID 22581228

  • Identification and Functional Characterisation of Novel Glucokinase Mutations Causing Maturity-Onset Diabetes of the Young in Slovakia PLOS ONE Valentinova, L., Beer, N. L., Stanik, J., Tribble, N. D., van de Bunt, M., Huckova, M., Barrett, A., Klimes, I., Gasperikova, D., Gloyn, A. L. 2012; 7 (4): e34541

    Abstract

    Heterozygous glucokinase (GCK) mutations cause a subtype of maturity-onset diabetes of the young (GCK-MODY). Over 600 GCK mutations have been reported of which ∼65% are missense. In many cases co-segregation has not been established and despite the importance of functional studies in ascribing pathogenicity for missense variants these have only been performed for <10% of mutations. The aim of this study was to determine the minimum prevalence of GCK-MODY amongst diabetic subjects in Slovakia by sequencing GCK in 100 Slovakian probands with a phenotype consistent with GCK-MODY and to explore the pathogenicity of identified variants through family and functional studies. Twenty-two mutations were identified in 36 families (17 missense) of which 7 (I110N, V200A, N204D, G258R, F419S, c.580-2A>C, c.1113-1114delGC) were novel. Parental DNA was available for 22 probands (covering 14/22 mutations) and co-segregation established in all cases. Bioinformatic analysis predicted all missense mutations to be damaging. Nine (I110N, V200A, N204D, G223S, G258R, F419S, V244G, L315H, I436N) mutations were functionally evaluated. Basic kinetic analysis explained pathogenicity for 7 mutants which showed reduced glucokinase activity with relative activity indices (RAI) between 0.6 to <0.001 compared to wild-type GCK (1.0). For the remaining 2 mutants additional molecular mechanisms were investigated. Differences in glucokinase regulatory protein (GKRP) -mediated-inhibition of GCK were observed for both L315H & I436N when compared to wild type (IC(50) 14.6±0.1 mM & 20.3±1.6 mM vs.13.3±0.1 mM respectively [p<0.03]). Protein instability as assessed by thermal lability studies demonstrated that both L315H and I436N show marked thermal instability compared to wild-type GCK (RAI at 55°C 8.8±0.8% & 3.1±0.4% vs. 42.5±3.9% respectively [p<0.001]). The minimum prevalence of GCK-MODY amongst Slovakian patients with diabetes was 0.03%. In conclusion, we have identified 22 GCK mutations in 36 Slovakian probands and demonstrate that combining family, bioinformatic and functional studies can aid the interpretation of variants identified by molecular diagnostic screening.

    View details for DOI 10.1371/journal.pone.0034541

    View details for Web of Science ID 000305012700044

    View details for PubMedID 22493702

    View details for PubMedCentralID PMC3321013

  • Novel Loci for Adiponectin Levels and Their Influence on Type 2 Diabetes and Metabolic Traits: A Multi-Ethnic Meta-Analysis of 45,891 Individuals PLOS GENETICS Dastani, Z., Hivert, M., Timpson, N., Perry, J. R., Yuan, X., Scott, R. A., Henneman, P., Heid, I. M., Kizer, J. R., Lyytikainen, L., Fuchsberger, C., Tanaka, T., Morris, A. P., Small, K., Isaacs, A., Beekman, M., Coassin, S., Lohman, K., Qi, L., Kanoni, S., Pankow, J. S., Uh, H., Wu, Y., Bidulescu, A., Rasmussen-Torvik, L. J., Greenwood, C. M., Ladouceur, M., Grimsby, J., Manning, A. K., Liu, C., Kooner, J., Mooser, V. E., Vollenweider, P., Kapur, K. A., Chambers, J., Wareham, N. J., Langenberg, C., Frants, R., Willems-Vandijk, K., Oostra, B. A., Willems, S. M., Lamina, C., Winkler, T. W., Psaty, B. M., Tracy, R. P., Brody, J., Chen, I., Viikari, J., Kahonen, M., Pramstaller, P. P., Evans, D. M., St Pourcain, B., Sattar, N., Wood, A. R., Bandinelli, S., Carlson, O. D., Egan, J. M., Bohringer, S., van Heemst, D., Kedenko, L., Kristiansson, K., Nuotio, M., Loo, B., Harris, T., Garcia, M., Kanaya, A., Haun, M., Klopp, N., Wichmann, H., Deloukas, P., Katsareli, E., Couper, D. J., Duncan, B. B., Kloppenburg, M., Adair, L. S., Borja, J. B., Wilson, J. G., Musani, S., Guo, X., Johnson, T., Semple, R., Teslovich, T. M., Allison, M. A., Redline, S., Buxbaum, S. G., Mohlke, K. L., Meulenbelt, I., Ballantyne, C. M., Dedoussis, G. V., Hu, F. B., Liu, Y., Paulweber, B., Spector, T. D., Slagboom, P. E., Ferrucci, L., Jula, A., Perola, M., Raitakari, O., Florez, J. C., Salomaa, V., Eriksson, J. G., Frayling, T. M., Hicks, A. A., Lehtimaki, T., Smith, G. D., Siscovick, D. S., Kronenberg, F., Van Duijn, C., Loos, R. J., Waterworth, D. M., Meigs, J. B., Dupuis, J., Richards, J. B. 2012; 8 (3)

    Abstract

    Circulating levels of adiponectin, a hormone produced predominantly by adipocytes, are highly heritable and are inversely associated with type 2 diabetes mellitus (T2D) and other metabolic traits. We conducted a meta-analysis of genome-wide association studies in 39,883 individuals of European ancestry to identify genes associated with metabolic disease. We identified 8 novel loci associated with adiponectin levels and confirmed 2 previously reported loci (P = 4.5×10(-8)-1.2×10(-43)). Using a novel method to combine data across ethnicities (N = 4,232 African Americans, N = 1,776 Asians, and N = 29,347 Europeans), we identified two additional novel loci. Expression analyses of 436 human adipocyte samples revealed that mRNA levels of 18 genes at candidate regions were associated with adiponectin concentrations after accounting for multiple testing (p<3×10(-4)). We next developed a multi-SNP genotypic risk score to test the association of adiponectin decreasing risk alleles on metabolic traits and diseases using consortia-level meta-analytic data. This risk score was associated with increased risk of T2D (p = 4.3×10(-3), n = 22,044), increased triglycerides (p = 2.6×10(-14), n = 93,440), increased waist-to-hip ratio (p = 1.8×10(-5), n = 77,167), increased glucose two hours post oral glucose tolerance testing (p = 4.4×10(-3), n = 15,234), increased fasting insulin (p = 0.015, n = 48,238), but with lower in HDL-cholesterol concentrations (p = 4.5×10(-13), n = 96,748) and decreased BMI (p = 1.4×10(-4), n = 121,335). These findings identify novel genetic determinants of adiponectin levels, which, taken together, influence risk of T2D and markers of insulin resistance.

    View details for DOI 10.1371/journal.pgen.1002607

    View details for Web of Science ID 000302254800080

  • Low-Frequency Variants in HMGA1 Are Not Associated With Type 2 Diabetes Risk DIABETES Marquez, M., Huyvaert, M., Perry, J. B., Pearson, R. D., Falchi, M., Morris, A. P., Vivequin, S., Lobbens, S., Yengo, L., Gaget, S., Pattou, F., Poulain-Godefroy, O., Charpentier, G., Carlsson, L. S., Jacobson, P., Sjostrom, L., Lantieri, O., Heude, B., Walley, A., Balkau, B., Marre, M., Froguel, P., Cauchi, S., DIAGRAM Consortium 2012; 61 (2): 524–30

    Abstract

    It has recently been suggested that the low-frequency c.136-14_136-13insC variant in high-mobility group A1 (HMGA1) may strongly contribute to insulin resistance and type 2 diabetes risk. In our study, we attempted to confirm that HMGA1 is a novel type 2 diabetes locus in French Caucasians. The gene was sequenced in 368 type 2 diabetic case subjects with a family history of type 2 diabetes and 372 normoglycemic control subjects without a family history of type 2 diabetes. None of the 41 genetic variations identified were associated with type 2 diabetes. The lack of association between the c.136-14_136-13insC variant and type 2 diabetes was confirmed in an independent French group of 4,538 case subjects and 4,015 control subjects and in a large meta-analysis of 16,605 case subjects and 46,179 control subjects. Finally, this variant had no effects on metabolic traits and was not involved in variations of HMGA1 and insulin receptor (INSR) expressions. The c.136-14_136-13insC variant was not associated with type 2 diabetes in individuals of European descent. Our study emphasizes the need to analyze a large number of subjects to reliably assess the association of low-frequency variants with the disease.

    View details for DOI 10.2337/db11-0728

    View details for Web of Science ID 000299798100031

    View details for PubMedID 22210315

    View details for PubMedCentralID PMC3266400

  • PTEN mutations as a cause of constitutive insulin sensitivity and obesity. The New England journal of medicine Pal, A. n., Barber, T. M., Van de Bunt, M. n., Rudge, S. A., Zhang, Q. n., Lachlan, K. L., Cooper, N. S., Linden, H. n., Levy, J. C., Wakelam, M. J., Walker, L. n., Karpe, F. n., Gloyn, A. L. 2012; 367 (11): 1002–11

    Abstract

    Epidemiologic and genetic evidence links type 2 diabetes, obesity, and cancer. The tumor-suppressor phosphatase and tensin homologue (PTEN) has roles in both cellular growth and metabolic signaling. Germline PTEN mutations cause a cancer-predisposition syndrome, providing an opportunity to study the effect of PTEN haploinsufficiency in humans.We measured insulin sensitivity and beta-cell function in 15 PTEN mutation carriers and 15 matched controls. Insulin signaling was measured in muscle and adipose-tissue biopsy specimens from 5 mutation carriers and 5 well-matched controls. We also assessed the effect of PTEN haploinsufficiency on obesity by comparing anthropometric indexes between the 15 patients and 2097 controls from a population-based study of healthy adults. Body composition was evaluated by means of dual-emission x-ray absorptiometry and skinfold thickness.Measures of insulin resistance were lower in the patients with a PTEN mutation than in controls (e.g., mean fasting plasma insulin level, 29 pmol per liter [range, 9 to 99] vs. 74 pmol per liter [range, 22 to 185]; P=0.001). This finding was confirmed with the use of hyperinsulinemic euglycemic clamping, showing a glucose infusion rate among carriers 2 times that among controls (P=0.009). The patients' insulin sensitivity could be explained by the presence of enhanced insulin signaling through the PI3K-AKT pathway, as evidenced by increased AKT phosphorylation. The PTEN mutation carriers were obese as compared with population-based controls (mean body-mass index [the weight in kilograms divided by the square of the height in meters], 32 [range, 23 to 42] vs. 26 [range, 15 to 48]; P<0.001). This increased body mass in the patients was due to augmented adiposity without corresponding changes in fat distribution.PTEN haploinsufficiency is a monogenic cause of profound constitutive insulin sensitization that is apparently obesogenic. We demonstrate an apparently divergent effect of PTEN mutations: increased risks of obesity and cancer but a decreased risk of type 2 diabetes owing to enhanced insulin sensitivity. (Funded by the Wellcome Trust and others.).

    View details for DOI 10.1056/NEJMoa1113966

    View details for PubMedID 22970944

    View details for PubMedCentralID PMC4072504

  • Meta-analysis of genome-wide association studies identifies eight new loci for type 2 diabetes in east Asians. Nature genetics Cho, Y. S., Chen, C., Hu, C., Long, J., Ong, R. T., Sim, X., Takeuchi, F., Wu, Y., Go, M. J., Yamauchi, T., Chang, Y., Kwak, S. H., Ma, R. C., Yamamoto, K., Adair, L. S., Aung, T., Cai, Q., Chang, L., Chen, Y., Gao, Y., Hu, F. B., Kim, H., Kim, S., Kim, Y. J., Lee, J. J., Lee, N. R., Li, Y., Liu, J. J., Lu, W., Nakamura, J., Nakashima, E., Ng, D. P., Tay, W. T., Tsai, F., Wong, T. Y., Yokota, M., Zheng, W., Zhang, R., Wang, C., So, W. Y., Ohnaka, K., Ikegami, H., Hara, K., Cho, Y. M., Cho, N. H., Chang, T., Bao, Y., Hedman, Å. K., Morris, A. P., McCarthy, M. I., Takayanagi, R., Park, K. S., Jia, W., Chuang, L., Chan, J. C., Maeda, S., Kadowaki, T., Lee, J., Wu, J., Teo, Y. Y., Tai, E. S., Shu, X. O., Mohlke, K. L., Kato, N., Han, B., Seielstad, M. 2012; 44 (1): 67-72

    Abstract

    We conducted a three-stage genetic study to identify susceptibility loci for type 2 diabetes (T2D) in east Asian populations. We followed our stage 1 meta-analysis of eight T2D genome-wide association studies (6,952 cases with T2D and 11,865 controls) with a stage 2 in silico replication analysis (5,843 cases and 4,574 controls) and a stage 3 de novo replication analysis (12,284 cases and 13,172 controls). The combined analysis identified eight new T2D loci reaching genome-wide significance, which mapped in or near GLIS3, PEPD, FITM2-R3HDML-HNF4A, KCNK16, MAEA, GCC1-PAX4, PSMD6 and ZFAND3. GLIS3, which is involved in pancreatic beta cell development and insulin gene expression, is known for its association with fasting glucose levels. The evidence of an association with T2D for PEPD and HNF4A has been shown in previous studies. KCNK16 may regulate glucose-dependent insulin secretion in the pancreas. These findings, derived from an east Asian population, provide new perspectives on the etiology of T2D.

    View details for DOI 10.1038/ng.1019

    View details for PubMedID 22158537

  • Insights into the pathogenicity of rare missense GCK variants from the identification and functional characterization of compound heterozygous and double mutations inherited in cis. Diabetes care Beer, N. L., Osbak, K. K., van de Bunt, M. n., Tribble, N. D., Steele, A. M., Wensley, K. J., Edghill, E. L., Colcough, K. n., Barrett, A. n., Valentínová, L. n., Rundle, J. K., Raimondo, A. n., Grimsby, J. n., Ellard, S. n., Gloyn, A. L. 2012; 35 (7): 1482–84

    Abstract

    To demonstrate the importance of using a combined genetic and functional approach to correctly interpret a genetic test for monogenic diabetes.We identified three probands with a phenotype consistent with maturity-onset diabetes of the young (MODY) subtype GCK-MODY, in whom two potential pathogenic mutations were identified: [R43H/G68D], [E248 K/I225M], or [G261R/D217N]. Allele-specific PCR and cosegregation were used to determine phase. Single and double mutations were kinetically characterized.The mutations occurred in cis (double mutants) in two probands and in trans in one proband. Functional studies of all double mutants revealed inactivating kinetics. The previously reported GCK-MODY mutations R43H and G68D were inherited from an affected father and unaffected mother, respectively. Both our functional and genetic studies support R43H as the cause of GCK-MODY and G68D as a neutral rare variant.These data highlight the need for family/functional studies, even for previously reported pathogenic mutations.

    View details for DOI 10.2337/dc11-2420

    View details for PubMedID 22611063

    View details for PubMedCentralID PMC3379612

  • Correlation of rare coding variants in the gene encoding human glucokinase regulatory protein with phenotypic, cellular, and kinetic outcomes. The Journal of clinical investigation Rees, M. G., Ng, D. n., Ruppert, S. n., Turner, C. n., Beer, N. L., Swift, A. J., Morken, M. A., Below, J. E., Blech, I. n., Mullikin, J. C., McCarthy, M. I., Biesecker, L. G., Gloyn, A. L., Collins, F. S. 2012; 122 (1): 205–17

    Abstract

    Defining the genetic contribution of rare variants to common diseases is a major basic and clinical science challenge that could offer new insights into disease etiology and provide potential for directed gene- and pathway-based prevention and treatment. Common and rare nonsynonymous variants in the GCKR gene are associated with alterations in metabolic traits, most notably serum triglyceride levels. GCKR encodes glucokinase regulatory protein (GKRP), a predominantly nuclear protein that inhibits hepatic glucokinase (GCK) and plays a critical role in glucose homeostasis. The mode of action of rare GCKR variants remains unexplored. We identified 19 nonsynonymous GCKR variants among 800 individuals from the ClinSeq medical sequencing project. Excluding the previously described common missense variant p.Pro446Leu, all variants were rare in the cohort. Accordingly, we functionally characterized all variants to evaluate their potential phenotypic effects. Defects were observed for the majority of the rare variants after assessment of cellular localization, ability to interact with GCK, and kinetic activity of the encoded proteins. Comparing the individuals with functional rare variants to those without such variants showed associations with lipid phenotypes. Our findings suggest that, while nonsynonymous GCKR variants, excluding p.Pro446Leu, are rare in individuals of mixed European descent, the majority do affect protein function. In sum, this study utilizes computational, cell biological, and biochemical methods to present a model for interpreting the clinical significance of rare genetic variants in common disease.

    View details for DOI 10.1172/JCI46425

    View details for PubMedID 22182842

    View details for PubMedCentralID PMC3248284

  • Cellular characterisation of the GCKR P446L variant associated with type 2 diabetes risk. Diabetologia Rees, M. G., Wincovitch, S. n., Schultz, J. n., Waterstradt, R. n., Beer, N. L., Baltrusch, S. n., Collins, F. S., Gloyn, A. L. 2012; 55 (1): 114–22

    Abstract

    Translation of genetic association signals into molecular mechanisms for diabetes has been slow. The glucokinase regulatory protein (GKRP; gene symbol GCKR) P446L variant, associated with inverse modulation of glucose- and lipid-related traits, has been shown to alter the kinetics of glucokinase (GCK) inhibition. As GCK inhibition is associated with nuclear sequestration, we aimed to determine whether this variant also alters the direct interaction between GKRP and GCK and their intracellular localisation.Fluorescently tagged rat and human wild-type (WT)- or P446L-GCKR and GCK were transiently transfected into HeLa cells and mouse primary hepatocytes. Whole-cell and nuclear fluorescence was quantified in individual cells exposed to low- or high-glucose conditions (5.5 or 25 mmol/l glucose, respectively). Interaction between GCK and GKRP was measured by sensitised emission-based fluorescence resonance energy transfer (FRET) efficiency.P446L-GKRP had a decreased degree of nuclear localisation, ability to sequester GCK and direct interaction with GCK as measured by FRET compared with WT-GKRP. Decreased interaction was observed between WT-GKRP and GCK at high compared with low glucose, but not between P446L-GKRP and GCK. Rat WT-GKRP and P446L-GKRP behaved quite differently: both variants responded to high glucose by diminished sequestration of GCK but showed no effect of the P446L variant on nuclear localisation or GCK sequestration.Our study suggests the common human P446L-GKRP variant protein results in elevated hepatic glucose uptake and disposal by increasing active cytosolic GCK. This would increase hepatic lipid biosynthesis but decrease fasting plasma glucose concentrations and provides a potential mechanism for the protective effect of this allele on type 2 diabetes risk.

    View details for DOI 10.1007/s00125-011-2348-5

    View details for PubMedID 22038520

    View details for PubMedCentralID PMC3276843

  • GLUT2 (SLC2A2) is not the principal glucose transporter in human pancreatic beta cells: Implications for understanding genetic association signals at this locus MOLECULAR GENETICS AND METABOLISM McCulloch, L. J., van de Bunt, M., Braun, M., Frayn, K. N., Clark, A., Gloyn, A. L. 2011; 104 (4): 648–53

    Abstract

    SLC2A2 encoding glucose transporter -2 (GLUT2) acts as the primary glucose transporter and sensor in rodent pancreatic islets and is widely assumed to play a similar role in humans. In healthy adults SLC2A2 variants are associated with elevated fasting plasma glucose (fpg) concentrations but physiological characterisation does not support a defect in pancreatic beta-cell function. Interspecies differences can create barriers for the follow up of disease association signals. We hypothesised that GLUT2 is not the principal glucose transporter in human beta-cells and that SLC2A2 variants exert their effect on fpg levels through defects in other tissues. SLC2A1-4 (GLUT 1-4) mRNA expression levels were determined in human and mouse islets, beta-cells, liver, muscle and adipose tissue by qRT-PCR whilst GLUT1-3 protein levels were examined by immunohistochemistry. The presence of all three glucose transporters was demonstrated in human and mouse islets and purified beta-cells. Quantitative expression profiling demonstrated that Slc2a2 is the predominant glucose transporter (expression >10 fold higher that Slc2a1) in mouse islets whilst SLC2A1 and SLC2A3 predominate in both human islets and beta-cells (expression 2.8 and 2.7 fold higher than SLC2A2 respectively). Our data therefore suggest that GLUT2 is unlikely to be the principal glucose transporter in human beta-cells and that SLC2A2 defects in other metabolic tissues drive the observed differences in glucose levels between carriers of SLC2A2 variants. Direct extrapolation from rodent to human islet glucose transporter activity is unlikely to be appropriate.

    View details for DOI 10.1016/j.ymgme.2011.08.026

    View details for Web of Science ID 000298021400033

    View details for PubMedID 21920790

  • A large multi-centre European study validates high-sensitivity C-reactive protein (hsCRP) as a clinical biomarker for the diagnosis of diabetes subtypes DIABETOLOGIA Thanabalasingham, G., Shah, N., Vaxillaire, M., Hansen, T., Tuomi, T., Gasperikova, D., Szopa, M., Tjora, E., James, T. J., Kokko, P., Loiseleur, F., Andersson, E., Gaget, S., Isomaa, B., Nowak, N., Raeder, H., Stanik, J., Njolstad, P. R., Malecki, M. T., Klimes, I., Groop, L., Pedersen, O., Froguel, P., McCarthy, M. I., Gloyn, A. L., Owen, K. R. 2011; 54 (11): 2801–10

    Abstract

    An accurate molecular diagnosis of diabetes subtype confers clinical benefits; however, many individuals with monogenic diabetes remain undiagnosed. Biomarkers could help to prioritise patients for genetic investigation. We recently demonstrated that high-sensitivity C-reactive protein (hsCRP) levels are lower in UK patients with hepatocyte nuclear factor 1 alpha (HNF1A)-MODY than in other diabetes subtypes. In this large multi-centre study we aimed to assess the clinical validity of hsCRP as a diagnostic biomarker, examine the genotype-phenotype relationship and compare different hsCRP assays.High-sensitivity CRP levels were analysed in individuals with HNF1A-MODY (n = 457), glucokinase (GCK)-MODY (n = 404), hepatocyte nuclear factor 4 alpha (HNF4A)-MODY (n = 54) and type 2 diabetes (n = 582) from seven European centres. Three common assays for hsCRP analysis were evaluated. We excluded 121 participants (8.1%) with hsCRP values >10 mg/l. The discriminative power of hsCRP with respect to diabetes aetiology was assessed by receiver operating characteristic curve-derived C-statistic.In all centres and irrespective of the assay method, meta-analysis confirmed significantly lower hsCRP levels in those with HNF1A-MODY than in those with other aetiologies (z score -21.8, p < 5 × 10(-105)). HNF1A-MODY cases with missense mutations had lower hsCRP levels than those with truncating mutations (0.03 vs 0.08 mg/l, p < 5 × 10(-5)). High-sensitivity CRP values between assays were strongly correlated (r (2) ≥ 0.91, p ≤ 1 × 10(-5)). Across the seven centres, the C-statistic for distinguishing HNF1A-MODY from young adult-onset type 2 diabetes ranged from 0.79 to 0.97, indicating high discriminative accuracy.In the largest study to date, we have established that hsCRP is a clinically valid biomarker for HNF1A-MODY in European populations. Given the modest costs and wide availability, hsCRP could translate rapidly into clinical practice, considerably improving diagnosis rates in monogenic diabetes.

    View details for DOI 10.1007/s00125-011-2261-y

    View details for Web of Science ID 000295679800009

    View details for PubMedID 21814873

  • Genome-wide association study identifies loci influencing concentrations of liver enzymes in plasma NATURE GENETICS Chambers, J. C., Zhang, W., Sehmi, J., Li, X., Wass, M. N., Van der Harst, P., Holm, H., Sanna, S., Kavousi, M., Baumeister, S. E., Coin, L. J., Deng, G., Gieger, C., Heard-Costa, N. L., Hottenga, J., Kuehnel, B., Kumar, V., Lagou, V., Liang, L., Luan, J., Vidal, P., Leach, I., O'Reilly, P. F., Peden, J. F., Rahmioglu, N., Soininen, P., Speliotes, E. K., Yuan, X., Thorleifsson, G., Alizadeh, B. Z., Atwood, L. D., Borecki, I. B., Brown, M. J., Charoen, P., Cucca, F., Das, D., de Geus, E. C., Dixon, A. L., Doering, A., Ehret, G., Eyjolfsson, G. I., Farrall, M., Forouhi, N. G., Friedrich, N., Goessling, W., Gudbjartsson, D. F., Harris, T. B., Hartikainen, A., Heath, S., Hirschfield, G. M., Hofman, A., Homuth, G., Hyppoenen, E., Janssen, H. A., Johnson, T., Kangas, A. J., Kema, I. P., Kuehn, J. P., Lai, S., Lathrop, M., Lerch, M. M., Li, Y., Liang, T., Lin, J., Loos, R. F., Martin, N. G., Moffatt, M. F., Montgomery, G. W., Munroe, P. B., Musunuru, K., Nakamura, Y., O'Donnell, C. J., Olafsson, I., Penninx, B. W., Pouta, A., Prins, B. P., Prokopenko, I., Puls, R., Ruokonen, A., Savolainen, M. J., Schlessinger, D., Schouten, J. L., Seedorf, U., Sen-Chowdhry, S., Siminovitch, K. A., Smit, J. H., Spector, T. D., Tan, W., Teslovich, T. M., Tukiainen, T., Uitterlinden, A. G., Van der Klauw, M. M., Vasan, R. S., Wallace, C., Wallaschofski, H., Wichmann, H., Willemsen, G., Wuertz, P., Xu, C., Yerges-Armstrong, L. M., Abecasis, G. R., Ahmadi, K. R., Boomsma, D. I., Caulfield, M., Cookson, W. O., van Duijn, C. M., Froguel, P., Matsuda, K., McCarthy, M. I., Meisinger, C., Mooser, V., Pietilainen, K. H., Schumann, G., Snieder, H., Sternberg, M. E., Stolk, R. P., Thomas, H. C., Thorsteinsdottir, U., Uda, M., Waeber, G., Wareham, N. J., Waterworth, D. M., Watkins, H., Whitfield, J. B., Witteman, J. M., Wolffenbuttel, B. R., Fox, C. S., Ala-Korpela, M., Stefansson, K., Vollenweider, P., Voelzke, H., Schadt, E. E., Scott, J., Jarvelin, M., Elliott, P., Kooner, J. S., Alcohol Genome-wide Assoc AlcGen, Diabet Genetics Replication, Genetic Invest Anthropometric, Global Lipids Genetics Consortium, Genetics Liver Dis GOLD Consortium, Int Consortium Blood Pressure ICBP, Meta-Anal Glucose Insulin-Related 2011; 43 (11): 1131–38

    Abstract

    Concentrations of liver enzymes in plasma are widely used as indicators of liver disease. We carried out a genome-wide association study in 61,089 individuals, identifying 42 loci associated with concentrations of liver enzymes in plasma, of which 32 are new associations (P = 10(-8) to P = 10(-190)). We used functional genomic approaches including metabonomic profiling and gene expression analyses to identify probable candidate genes at these regions. We identified 69 candidate genes, including genes involved in biliary transport (ATP8B1 and ABCB11), glucose, carbohydrate and lipid metabolism (FADS1, FADS2, GCKR, JMJD1C, HNF1A, MLXIPL, PNPLA3, PPP1R3B, SLC2A2 and TRIB1), glycoprotein biosynthesis and cell surface glycobiology (ABO, ASGR1, FUT2, GPLD1 and ST3GAL4), inflammation and immunity (CD276, CDH6, GCKR, HNF1A, HPR, ITGA1, RORA and STAT4) and glutathione metabolism (GSTT1, GSTT2 and GGT), as well as several genes of uncertain or unknown function (including ABHD12, EFHD1, EFNA1, EPHA2, MICAL3 and ZNF827). Our results provide new insight into genetic mechanisms and pathways influencing markers of liver function.

    View details for DOI 10.1038/ng.970

    View details for Web of Science ID 000296584000019

    View details for PubMedID 22001757

    View details for PubMedCentralID PMC3482372

  • Genome-Wide Association Identifies Nine Common Variants Associated With Fasting Proinsulin Levels and Provides New Insights Into the Pathophysiology of Type 2 Diabetes DIABETES Strawbridge, R. J., Dupuis, J., Prokopenko, I., Barker, A., Ahlqvist, E., Rybin, D., Petrie, J. R., Travers, M. E., Bouatia-Naji, N., Dimas, A. S., Nica, A., Wheeler, E., Chen, H., Voight, B. F., Taneera, J., Kanoni, S., Peden, J. F., Turrini, F., Gustafsson, S., Zabena, C., Almgren, P., Barker, D. J., Barnes, D., Dennison, E. M., Eriksson, J. G., Eriksson, P., Eury, E., Folkersen, L., Fox, C. S., Frayling, T. M., Goel, A., Gu, H. F., Horikoshi, M., Isomaa, B., Jackson, A. U., Jameson, K. A., Kajantie, E., Kerr-Conte, J., Kuulasmaa, T., Kuusisto, J., Loos, R. J., Luan, J., Makrilakis, K., Manning, A. K., Teresa Martinez-Larrad, M., Narisu, N., Mannila, M. N., Ohrvik, J., Osmond, C., Pascoe, L., Payne, F., Sayer, A. A., Sennblad, B., Silveira, A., Stancakova, A., Stirrups, K., Swift, A. J., Syvanen, A., Tuomi, T., van 't Hooft, F. M., Walker, M., Weedon, M. N., Xie, W., Zethelius, B., Ongen, H., Malarstig, A., Hopewell, J. C., Saleheen, D., Chambers, J., Parish, S., Danesh, J., Kooner, J., Ostenson, C., Lind, L., Cooper, C. C., Serrano-Rios, M., Ferrannini, E., Forsen, T. J., Clarke, R., Franzosi, M. G., Seedorf, U., Watkins, H., Froguel, P., Johnson, P., Deloukas, P., Collins, F. S., Laakso, M., Dermitzakis, E. T., Boehnke, M., McCarthy, M. I., Wareham, N. J., Groop, L., Pattou, F., Gloyn, A. L., Dedoussis, G. V., Lyssenko, V., Meigs, J. B., Barroso, I., Watanabe, R. M., Ingelsson, E., Langenberg, C., Hamsten, A., Florez, J. C. 2011; 60 (10): 2624-2634

    Abstract

    Proinsulin is a precursor of mature insulin and C-peptide. Higher circulating proinsulin levels are associated with impaired β-cell function, raised glucose levels, insulin resistance, and type 2 diabetes (T2D). Studies of the insulin processing pathway could provide new insights about T2D pathophysiology.We have conducted a meta-analysis of genome-wide association tests of ∼2.5 million genotyped or imputed single nucleotide polymorphisms (SNPs) and fasting proinsulin levels in 10,701 nondiabetic adults of European ancestry, with follow-up of 23 loci in up to 16,378 individuals, using additive genetic models adjusted for age, sex, fasting insulin, and study-specific covariates.Nine SNPs at eight loci were associated with proinsulin levels (P < 5 × 10(-8)). Two loci (LARP6 and SGSM2) have not been previously related to metabolic traits, one (MADD) has been associated with fasting glucose, one (PCSK1) has been implicated in obesity, and four (TCF7L2, SLC30A8, VPS13C/C2CD4A/B, and ARAP1, formerly CENTD2) increase T2D risk. The proinsulin-raising allele of ARAP1 was associated with a lower fasting glucose (P = 1.7 × 10(-4)), improved β-cell function (P = 1.1 × 10(-5)), and lower risk of T2D (odds ratio 0.88; P = 7.8 × 10(-6)). Notably, PCSK1 encodes the protein prohormone convertase 1/3, the first enzyme in the insulin processing pathway. A genotype score composed of the nine proinsulin-raising alleles was not associated with coronary disease in two large case-control datasets.We have identified nine genetic variants associated with fasting proinsulin. Our findings illuminate the biology underlying glucose homeostasis and T2D development in humans and argue against a direct role of proinsulin in coronary artery disease pathogenesis.

    View details for DOI 10.2337/db11-0415

    View details for Web of Science ID 000295998700022

    View details for PubMedID 21873549

    View details for PubMedCentralID PMC3178302

  • The previously reported T342P GCK missense variant is not a pathogenic mutation causing MODY DIABETOLOGIA Steele, A. M., Tribble, N. D., Caswell, R., Wensley, K. J., Hattersley, A. T., Gloyn, A. L., Ellard, S. 2011; 54 (8): 2202–5

    View details for DOI 10.1007/s00125-011-2194-5

    View details for Web of Science ID 000292562500030

    View details for PubMedID 21604084

  • High-Sensitivity CRP Discriminates HNF1A-MODY From Other Subtypes of Diabetes DIABETES CARE McDonald, T. J., Shields, B. M., Lawry, J., Owen, K. R., Gloyn, A. L., Ellard, S., Hattersley, A. T. 2011; 34 (8): 1860–62

    Abstract

    Maturity-onset diabetes of the young (MODY) as a result of mutations in hepatocyte nuclear factor 1-α (HNF1A) is often misdiagnosed as type 1 diabetes or type 2 diabetes. Recent work has shown that high-sensitivity C-reactive protein (hs-CRP) levels are lower in HNF1A-MODY than type 1 diabetes, type 2 diabetes, or glucokinase (GCK)-MODY. We aim to replicate these findings in larger numbers and other MODY subtypes.hs-CRP levels were assessed in 750 patients (220 HNF1A, 245 GCK, 54 HNF4-α [HNF4A], 21 HNF1-β (HNF1B), 53 type 1 diabetes, and 157 type 2 diabetes).hs-CRP was lower in HNF1A-MODY (median [IQR] 0.3 [0.1-0.6] mg/L) than type 2 diabetes (1.40 [0.60-3.45] mg/L; P < 0.001) and type 1 diabetes (1.10 [0.50-1.85] mg/L; P < 0.001), HNF4A-MODY (1.45 [0.46-2.88] mg/L; P < 0.001), GCK-MODY (0.60 [0.30-1.80] mg/L; P < 0.001), and HNF1B-MODY (0.60 [0.10-2.8] mg/L; P = 0.07). hs-CRP discriminated HNF1A-MODY from type 2 diabetes with hs-CRP <0.75 mg/L showing 79% sensitivity and 70% specificity (receiver operating characteristic area under the curve = 0.84).hs-CRP levels are lower in HNF1A-MODY than other forms of diabetes and may be used as a biomarker to select patients for diagnostic HNF1A genetic testing.

    View details for DOI 10.2337/dc11-0323

    View details for Web of Science ID 000294035400034

    View details for PubMedID 21700917

    View details for PubMedCentralID PMC3142017

  • Identification of an imprinted master trans regulator at the KLF14 locus related to multiple metabolic phenotypes NATURE GENETICS Small, K. S., Hedman, A. K., Grundberg, E., Nica, A. C., Thorleifsson, G., Kong, A., Thorsteindottir, U., Shin, S., Richards, H. B., Soranzo, N., Ahmadi, K. R., Lindgren, C. M., Stefansson, K., Dermitzakis, E. T., Deloukas, P., Spector, T. D., McCarthy, M. I., GIANT Consortium, MAGIC Investigators, DIAGRAM Consortium, MuTHER Consortium 2011; 43 (6): 561–U90

    Abstract

    Genome-wide association studies have identified many genetic variants associated with complex traits. However, at only a minority of loci have the molecular mechanisms mediating these associations been characterized. In parallel, whereas cis regulatory patterns of gene expression have been extensively explored, the identification of trans regulatory effects in humans has attracted less attention. Here we show that the type 2 diabetes and high-density lipoprotein cholesterol-associated cis-acting expression quantitative trait locus (eQTL) of the maternally expressed transcription factor KLF14 acts as a master trans regulator of adipose gene expression. Expression levels of genes regulated by this trans-eQTL are highly correlated with concurrently measured metabolic traits, and a subset of the trans-regulated genes harbor variants directly associated with metabolic phenotypes. This trans-eQTL network provides a mechanistic understanding of the effect of the KLF14 locus on metabolic disease risk and offers a potential model for other complex traits.

    View details for DOI 10.1038/ng.833

    View details for Web of Science ID 000291017000013

    View details for PubMedID 21572415

    View details for PubMedCentralID PMC3192952

  • Comprehensive Human Adipose Tissue mRNA and MicroRNA Endogenous Control Selection for Quantitative Real-Time-PCR Normalization OBESITY Neville, M. J., Collins, J. M., Gloyn, A. L., McCarthy, M. I., Karpe, F. 2011; 19 (4): 888–92

    Abstract

    The accurate quantification of cellular and tissue mRNA and microRNA content is reliant upon the selection of stable endogenous control transcripts for normalizing quantitative real-time-PCR (qRT-PCR) data. Using the combination of unbiased and informed approaches and a wide range of human adipose tissues and cells, we sought to identify invariant control transcripts for mRNA and microRNA. A total of 26 mRNA transcript candidates were selected from the literature. MicroRNA candidates were selected from a microRNA-microarray (Agilent, n = 22 tissues), and together with candidates from the literature resulted in 14 different microRNAs. The variability of these mRNA and microRNA transcripts were then tested in a large (n = 180) collection of a variety of human adipose tissues and cell samples. Phosphoglycerate kinase-1 (PGK1) and peptidylprolyl isomerase A (PPIA) were identified as the most stable mRNAs across all tissues and panels. MiR-103 was overall the most stable microRNA transcript across all biological backgrounds. Several proposed and commonly used normalization transcripts were found to be highly variable. We then tested the effect on expression of two established adipocyte-related transcripts (fatty acid binding protein 4 (FABP4) and microRNA-145 (miR-145)), either normalized to the optimal or a commonly used controls transcript. This test clearly indicated that spurious results could arise from using less stable control transcripts for mRNA and microRNA qRT-PCR.

    View details for DOI 10.1038/oby.2010.257

    View details for Web of Science ID 000288901400034

    View details for PubMedID 20948521

    View details for PubMedCentralID PMC4623139

  • Discovery of a novel site regulating glucokinase activity following characterization of a new mutation causing hyperinsulinemic hypoglycemia in humans. The Journal of biological chemistry Beer, N. L., van de Bunt, M. n., Colclough, K. n., Lukacs, C. n., Arundel, P. n., Chik, C. L., Grimsby, J. n., Ellard, S. n., Gloyn, A. L. 2011; 286 (21): 19118–26

    Abstract

    Type 2 diabetes is a global problem, and current ineffective therapeutic strategies pave the way for novel treatments like small molecular activators targeting glucokinase (GCK). GCK activity is fundamental to beta cell and hepatocyte glucose metabolism, and heterozygous activating and inactivating GCK mutations cause hyperinsulinemic hypoglycemia (HH) and maturity onset diabetes of the young (MODY) respectively. Over 600 naturally occurring inactivating mutations have been reported, whereas only 13 activating mutations are documented to date. We report two novel GCK HH mutations (V389L and T103S) at residues where MODY mutations also occur (V389D and T103I). Using recombinant proteins with in vitro assays, we demonstrated that both HH mutants had a greater relative activity index than wild type (6.0 for V389L, 8.4 for T103S, and 1.0 for wild type). This was driven by an increased affinity for glucose (S(0.5), 3.3 ± 0.1 and 3.5 ± 0.1 mm, respectively) versus wild type (7.5 ± 0.1 mm). Correspondingly, the V389D and T103I MODY mutants had markedly reduced relative activity indexes (<0.1). T103I had an altered affinity for glucose (S(0.5), 24.9 ± 0.6 mm), whereas V389D also exhibited a reduced affinity for ATP and decreased catalysis rate (S(0.5), 78.6 ± 4.5 mm; ATP(K(m)), 1.5 ± 0.1 mm; K(cat), 10.3 ± 1.1s(-1)) compared with wild type (ATP(K(m)), 0.4 ± <0.1; K(cat), 62.9 ± 1.2). Both Thr-103 mutants showed reduced inhibition by the endogenous hepatic inhibitor glucokinase regulatory protein. Molecular modeling demonstrated that Thr-103 maps to the allosteric activator site, whereas Val-389 is located remotely to this position and all other previously reported activating mutations, highlighting α-helix 11 as a novel region regulating GCK activity. Our data suggest that pharmacological manipulation of GCK activity at locations distal from the allosteric activator site is possible.

    View details for DOI 10.1074/jbc.M111.223362

    View details for PubMedID 21454522

    View details for PubMedCentralID PMC3099725

  • Genetically Programmed Defects in beta-Cell Function BETASYS: SYSTEMS BIOLOGY OF REGULATED EXOCYTOSIS IN PANCREATIC BETA-CELLS Pal, A., Gloyn, A. L., BoossBavnbek, B., Klosgen, B., Larsen, J., Pociot, F., Renstrom, E. 2011; 2: 299–326
  • A role for coding functional variants in HNF4A in type 2 diabetes susceptibility DIABETOLOGIA Jafar-Mohammadi, B., Groves, C. J., Gjesing, A. P., Herrera, B. M., Winckler, W., Stringham, H. M., Morris, A. P., Lauritzen, T., Doney, A. F., Morris, A. D., Weedon, M. N., Swift, A. J., Kuusisto, J., Laakso, M., Altshuler, D., Hattersley, A. T., Collins, F. S., Boehnke, M., Hansen, T., Pedersen, O., Palmer, C. A., Frayling, T. M., Gloyn, A. L., McCarthy, M. I., DIAGRAM Consortium 2011; 54 (1): 111–19

    Abstract

    Rare mutations in the gene HNF4A, encoding the transcription factor hepatocyte nuclear factor 4α (HNF-4A), account for ~5% of cases of MODY and more frequent variants in this gene may be involved in multifactorial forms of diabetes. Two low-frequency, non-synonymous variants in HNF4A (V255M, minor allele frequency [MAF] ~0.1%; T130I, MAF ~3.0%)-known to influence downstream HNF-4A target gene expression-are of interest, but previous type 2 diabetes association reports were inconclusive. We aimed to evaluate the contribution of these variants to type 2 diabetes susceptibility through large-scale association analysis.We genotyped both variants in at least 5,745 cases and 14,756 population controls from the UK and Denmark. We also undertook an expanded association analysis that included previously reported and novel genotype data obtained in Danish, Finnish, Canadian and Swedish samples. A meta-analysis incorporating all published association studies of the T130I variant was subsequently carried out in a maximum sample size of 14,279 cases and 26,835 controls.We found no association between V255M and type 2 diabetes in either the initial (p = 0.28) or the expanded analysis (p = 0.44). However, T130I demonstrated a modest association with type 2 diabetes in the UK and Danish samples (additive per allele OR 1.17 [95% CI 1.08-1.28]; p = 1.5 × 10⁻⁴), which was strengthened in the meta-analysis (OR 1.20 [95% CI 1.10-1.30]; p = 2.1 × 10⁻⁵).Our data are consistent with T130I as a low-frequency variant influencing type 2 diabetes risk, but are not conclusive when judged against stringent standards for genome-wide significance. This study exemplifies the difficulties encountered in association testing of low-frequency variants.

    View details for DOI 10.1007/s00125-010-1916-4

    View details for Web of Science ID 000284896900018

    View details for PubMedID 20878384

    View details for PubMedCentralID PMC3119815

  • From Genetic Association to Molecular Mechanism CURRENT DIABETES REPORTS van de Bunt, M., Gloyn, A. L. 2010; 10 (6): 452–66

    Abstract

    Over the past 3 years, there has been a dramatic increase in the number of confirmed type 2 diabetes (T2D) susceptibility loci, most arising through the implementation of genome-wide association studies (GWAS). However, progress toward the understanding of disease mechanisms has been slowed by modest effect sizes and the fact that most GWAS signals map away from coding sequence: the presumption is that their effects are mediated through regulation of nearby transcripts, but the identities of the genes concerned are often far from clear. In this review we describe the progress that has been made to date in translating association signals into molecular mechanisms with a focus on the most tractable signals (eg, KCNJ11/ABCC8, SLC30A8, GCKR) and those in which human, animal, and cellular models (FTO, TCF7L2, G6PC2) have provided insights into the role in T2D pathogenesis. Finally, the challenges for the field with the advent of genome-scale next-generation resequencing efforts are discussed.

    View details for DOI 10.1007/s11892-010-0150-2

    View details for Web of Science ID 000288495200007

    View details for PubMedID 20878272

  • Variation across the allele frequency spectrum NATURE GENETICS Gloyn, A. L., McCarthy, M. I. 2010; 42 (8): 648–50

    View details for DOI 10.1038/ng0810-648

    View details for Web of Science ID 000280524000004

    View details for PubMedID 20664646

  • Twelve type 2 diabetes susceptibility loci identified through large-scale association analysis NATURE GENETICS Voight, B. F., Scott, L. J., Steinthorsdottir, V., Morris, A. P., Dina, C., Welch, R. P., Zeggini, E., Huth, C., Aulchenko, Y. S., Thorleifsson, G., McCulloch, L. J., Ferreira, T., Grallert, H., Amin, N., Wu, G., Willer, C. J., Raychaudhuri, S., McCarroll, S. A., Langenberg, C., Hofmann, O. M., Dupuis, J., Qi, L., Segre, A. V., van Hoek, M., Navarro, P., Ardlie, K., Balkau, B., Benediktsson, R., Bennett, A. J., Blagieva, R., Boerwinkle, E., Bonnycastle, L. L., Bostrom, K. B., Bravenboer, B., Bumpstead, S., Burtt, N. P., Charpentier, G., Chines, P. S., Cornelis, M., Couper, D. J., Crawford, G., Doney, A. S., Elliott, K. S., Elliott, A. L., Erdos, M. R., Fox, C. S., Franklin, C. S., Ganser, M., Gieger, C., Grarup, N., Green, T., Griffin, S., Groves, C. J., Guiducci, C., Hadjadj, S., Hassanali, N., Herder, C., Isomaa, B., Jackson, A. U., Johnson, P. R., Jorgensen, T., Kao, W. H., Klopp, N., Kong, A., Kraft, P., Kuusisto, J., Lauritzen, T., Li, M., Lieverse, A., Lindgren, C. M., Lyssenko, V., Marre, M., Meitinger, T., Midthjell, K., Morken, M. A., Narisu, N., Nilsson, P., Owen, K. R., Payne, F., Perry, J. R., Petersen, A., Platou, C., Proenca, C., Prokopenko, I., Rathmann, W., Rayner, N. W., Robertson, N. R., Rocheleau, G., Roden, M., Sampson, M. J., Saxena, R., Shields, B. M., Shrader, P., Sigurdsson, G., Sparso, T., Strassburger, K., Stringham, H. M., Sun, Q., Swift, A. J., Thorand, B., Tichet, J., Tuomi, T., van Dam, R. M., Van Haeften, T. W., van Herpt, T., van Vliet-Ostaptchouk, J. V., Walters, G. B., Weedon, M. N., Wijmenga, C., Witteman, J., Bergman, R. N., Cauchi, S., Collins, F. S., Gloyn, A. L., Gyllensten, U., Hansen, T., Hide, W. A., Hitman, G. A., Hofman, A., Hunter, D. J., Hveem, K., Laakso, M., Mohlke, K. L., Morris, A. D., Palmer, C. N., Pramstaller, P. P., Rudan, I., Sijbrands, E., Stein, L. D., Tuomilehto, J., Uitterlinden, A., Walker, M., Wareham, N. J., Watanabe, R. M., Abecasis, G. R., Boehm, B. O., Campbell, H., Daly, M. J., Hattersley, A. T., Hu, F. B., Meigs, J. B., Pankow, J. S., Pedersen, O., Wichmann, H., Barroso, I., Florez, J. C., Frayling, T. M., Groop, L., Sladek, R., Thorsteinsdottir, U., Wilson, J. F., Illig, T., Froguel, P., van Duijn, C. M., Stefansson, K., Altshuler, D., Boehnke, M., McCarthy, M. I. 2010; 42 (7): 579-U155

    Abstract

    By combining genome-wide association data from 8,130 individuals with type 2 diabetes (T2D) and 38,987 controls of European descent and following up previously unidentified meta-analysis signals in a further 34,412 cases and 59,925 controls, we identified 12 new T2D association signals with combined P<5x10(-8). These include a second independent signal at the KCNQ1 locus; the first report, to our knowledge, of an X-chromosomal association (near DUSP9); and a further instance of overlap between loci implicated in monogenic and multifactorial forms of diabetes (at HNF1A). The identified loci affect both beta-cell function and insulin action, and, overall, T2D association signals show evidence of enrichment for genes involved in cell cycle regulation. We also show that a high proportion of T2D susceptibility loci harbor independent association signals influencing apparently unrelated complex traits.

    View details for DOI 10.1038/ng.609

    View details for Web of Science ID 000279242400010

    View details for PubMedID 20581827

    View details for PubMedCentralID PMC3080658

  • Global microRNA expression profiles in insulin target tissues in a spontaneous rat model of type 2 diabetes DIABETOLOGIA Herrera, B. M., Lockstone, H. E., Taylor, J. M., Ria, M., Barrett, A., Collins, S., Kaisaki, P., Argoud, K., Fernandez, C., Travers, M. E., Grew, J. P., Randall, J. C., Gloyn, A. L., Gauguier, D., McCarthy, M. I., Lindgren, C. M. 2010; 53 (6): 1099–1109

    Abstract

    MicroRNAs regulate a broad range of biological mechanisms. To investigate the relationship between microRNA expression and type 2 diabetes, we compared global microRNA expression in insulin target tissues from three inbred rat strains that differ in diabetes susceptibility.Using microarrays, we measured the expression of 283 microRNAs in adipose, liver and muscle tissue from hyperglycaemic (Goto-Kakizaki), intermediate glycaemic (Wistar Kyoto) and normoglycaemic (Brown Norway) rats (n = 5 for each strain). Expression was compared across strains and validated using quantitative RT-PCR. Furthermore, microRNA expression variation in adipose tissue was investigated in 3T3-L1 adipocytes exposed to hyperglycaemic conditions.We found 29 significantly differentiated microRNAs (p(adjusted) < 0.05): nine in adipose tissue, 18 in liver and two in muscle. Of these, five microRNAs had expression patterns that correlated with the strain-specific glycaemic phenotype. MiR-222 (p(adjusted) = 0.0005) and miR-27a (p(adjusted) = 0.006) were upregulated in adipose tissue; miR-195 (p(adjusted) = 0.006) and miR-103 (p(adjusted) = 0.04) were upregulated in liver; and miR-10b (p(adjusted) = 0.004) was downregulated in muscle. Exposure of 3T3-L1 adipocytes to increased glucose concentration upregulated the expression of miR-222 (p = 0.008), miR-27a (p = 0.02) and the previously reported miR-29a (p = 0.02). Predicted target genes of these differentially expressed microRNAs are involved in pathways relevant to type 2 diabetes.The expression patterns of miR-222, miR-27a, miR-195, miR-103 and miR-10b varied with hyperglycaemia, suggesting a role for these microRNAs in the pathophysiology of type 2 diabetes, as modelled by the Gyoto-Kakizaki rat. We observed similar patterns of expression of miR-222, miR-27a and miR-29a in adipocytes as a response to increased glucose levels, which supports our hypothesis that altered expression of microRNAs accompanies primary events related to the pathogenesis of type 2 diabetes.

    View details for DOI 10.1007/s00125-010-1667-2

    View details for Web of Science ID 000277138100012

    View details for PubMedID 20198361

    View details for PubMedCentralID PMC2860560

  • New genetic loci implicated in fasting glucose homeostasis and their impact on type 2 diabetes risk NATURE GENETICS Dupuis, J., Langenberg, C., Prokopenko, I., Saxena, R., Soranzo, N., Jackson, A. U., Wheeler, E., Glazer, N. L., Bouatia-Naji, N., Gloyn, A. L., Lindgren, C. M., Magi, R., Morris, A. P., Randall, J., Johnson, T., Elliott, P., Rybin, D., Thorleifsson, G., Steinthorsdottir, V., Henneman, P., Grallert, H., Dehghan, A., Hottenga, J. J., Franklin, C. S., Navarro, P., Song, K., Goel, A., Perry, J. R., Egan, J. M., Lajunen, T., Grarup, N., Sparso, T., Doney, A., Voight, B. F., Stringham, H. M., Li, M., Kanoni, S., Shrader, P., Cavalcanti-Proenca, C., Kumari, M., Qi, L., Timpson, N. J., Gieger, C., Zabena, C., Rocheleau, G., Ingelsson, E., An, P., O'Connell, J., Luan, J., Elliott, A., McCarroll, S. A., Payne, F., Roccasecca, R. M., Pattou, F., Sethupathy, P., Ardlie, K., Ariyurek, Y., Balkau, B., Barter, P., Beilby, J. P., Ben-Shlomo, Y., Benediktsson, R., Bennett, A. J., Bergmann, S., Bochud, M., Boerwinkle, E., Bonnefond, A., Bonnycastle, L. L., Borch-Johnsen, K., Boettcher, Y., Brunner, E., Bumpstead, S. J., Charpentier, G., Chen, Y. I., Chines, P., Clarke, R., Coin, L. J., Cooper, M. N., Cornelis, M., Crawford, G., Crisponi, L., Day, I. N., de Geus, E. J., Delplanque, J., Dina, C., Erdos, M. R., Fedson, A. C., Fischer-Rosinsky, A., Forouhi, N. G., Fox, C. S., Frants, R., Franzosi, M. G., Galan, P., Goodarzi, M. O., Graessler, J., Groves, C. J., Grundy, S., Gwilliam, R., Gyllensten, U., Hadjadj, S., Hallmans, G., Hammond, N., Han, X., Hartikainen, A., Hassanali, N., Hayward, C., Heath, S. C., Hercberg, S., Herder, C., Hicks, A. A., Hillman, D. R., Hingorani, A. D., Hofman, A., Hui, J., Hung, J., Isomaa, B., Johnson, P. R., Jorgensen, T., Jula, A., Kaakinen, M., Kaprio, J., Kesaniemi, Y. A., Kivimaki, M., Knight, B., Koskinen, S., Kovacs, P., Kyvik, K. O., Lathrop, G. M., Lawlor, D. A., Le Bacquer, O., Lecoeur, C., Li, Y., Lyssenko, V., Mahley, R., Mangino, M., Manning, A. K., Teresa Martinez-Larrad, M., McAteer, J. B., McCulloch, L. J., McPherson, R., Meisinger, C., Melzer, D., Meyre, D., Mitchell, B. D., Morken, M. A., Mukherjee, S., Naitza, S., Narisu, N., Neville, M. J., Oostra, B. A., Orru, M., Pakyz, R., Palmer, C. N., Paolisso, G., Pattaro, C., Pearson, D., Peden, J. F., Pedersen, N. L., Perola, M., Pfeiffer, A. F., Pichler, I., Polasek, O., Posthuma, D., Potter, S. C., Pouta, A., Province, M. A., Psaty, B. M., Rathmann, W., Rayner, N. W., Rice, K., Ripatti, S., Rivadeneira, F., Roden, M., Rolandsson, O., Sandbaek, A., Sandhu, M., Sanna, S., Sayer, A. A., Scheet, P., Scott, L. J., Seedorf, U., Sharp, S. J., Shields, B., Sigurosson, G., Sijbrands, E. J., Silveira, A., Simpson, L., Singleton, A., Smith, N. L., Sovio, U., Swift, A., Syddall, H., Syvanen, A., Tanaka, T., Thorand, B., Tichet, J., Tonjes, A., Tuomi, T., Uitterlinden, A. G., Van Dijk, K. W., van Hoek, M., Varma, D., Visvikis-Siest, S., Vitart, V., Vogelzangs, N., Waeber, G., Wagner, P. J., Walley, A., Walters, G. B., Ward, K. L., Watkins, H., Weedon, M. N., Wild, S. H., Willemsen, G., Witteman, J. C., Yarnell, J. W., Zeggini, E., Zelenika, D., Zethelius, B., Zhai, G., Zhao, J. H., Zillikens, M. C., Borecki, I. B., Loos, R. J., Meneton, P., Magnusson, P. K., Nathan, D. M., Williams, G. H., Hattersley, A. T., Silander, K., Salomaa, V., Smith, G. D., Bornstein, S. R., Schwarz, P., Spranger, J., Karpe, F., Shuldiner, A. R., Cooper, C., Dedoussis, G. V., Serrano-Rios, M., Morris, A. D., Lind, L., Palmer, L. J., Hu, F. B., Franks, P. W., Ebrahim, S., Marmot, M., Kao, W. H., Pankow, J. S., Sampson, M. J., Kuusisto, J., Laakso, M., Hansen, T., Pedersen, O., Pramstaller, P. P., Wichmann, H. E., Illig, T., Rudan, I., Wright, A. F., Stumvoll, M., Campbell, H., Wilson, J. F., Bergman, R. N., Buchanan, T. A., Collins, F. S., Mohlke, K. L., Tuomilehto, J., Valle, T. T., Altshuler, D., Rotter, J. I., Siscovick, D. S., Penninx, B. W., Boomsma, D. I., Deloukas, P., Spector, T. D., Frayling, T. M., Ferrucci, L., Kong, A., Thorsteinsdottir, U., Stefansson, K., van Duijn, C. M., Aulchenko, Y. S., Cao, A., Scuteri, A., Schlessinger, D., Uda, M., Ruokonen, A., Jarvelin, M., Waterworth, D. M., Vollenweider, P., Peltonen, L., Mooser, V., Abecasis, G. R., Wareham, N. J., Sladek, R., Froguel, P., Watanabe, R. M., Meigs, J. B., Groop, L., Boehnke, M., McCarthy, M. I., Florez, J. C., Barroso, I. 2010; 42 (2): 105-U32

    Abstract

    Levels of circulating glucose are tightly regulated. To identify new loci influencing glycemic traits, we performed meta-analyses of 21 genome-wide association studies informative for fasting glucose, fasting insulin and indices of beta-cell function (HOMA-B) and insulin resistance (HOMA-IR) in up to 46,186 nondiabetic participants. Follow-up of 25 loci in up to 76,558 additional subjects identified 16 loci associated with fasting glucose and HOMA-B and two loci associated with fasting insulin and HOMA-IR. These include nine loci newly associated with fasting glucose (in or near ADCY5, MADD, ADRA2A, CRY2, FADS1, GLIS3, SLC2A2, PROX1 and C2CD4B) and one influencing fasting insulin and HOMA-IR (near IGF1). We also demonstrated association of ADCY5, PROX1, GCK, GCKR and DGKB-TMEM195 with type 2 diabetes. Within these loci, likely biological candidate genes influence signal transduction, cell proliferation, development, glucose-sensing and circadian regulation. Our results demonstrate that genetic studies of glycemic traits can identify type 2 diabetes risk loci, as well as loci containing gene variants that are associated with a modest elevation in glucose levels but are not associated with overt diabetes.

    View details for DOI 10.1038/ng.520

    View details for Web of Science ID 000274084400005

    View details for PubMedID 20081858

  • Evaluation of Serum 1,5 Anhydroglucitol Levels as a Clinical Test to Differentiate Subtypes of Diabetes DIABETES CARE Pal, A., Farmer, A. J., Dudley, C., Selwood, M. P., Barrow, B. A., Klyne, R., Grew, J. P., McCarthy, M. I., Gloyn, A. L., Owen, K. R. 2010; 33 (2): 252–57

    Abstract

    Assignment of the correct molecular diagnosis in diabetes is necessary for informed decisions regarding treatment and prognosis. Better clinical markers would facilitate discrimination and prioritization for genetic testing between diabetes subtypes. Serum 1,5 anhydroglucitol (1,5AG) levels were reported to differentiate maturity-onset diabetes of the young due to HNF1A mutations (HNF1A-MODY) from type 2 diabetes, but this requires further validation. We evaluated serum 1,5AG in a range of diabetes subtypes as an adjunct for defining diabetes etiology.1,5AG was measured in U.K. subjects with: HNF1A-MODY (n = 23), MODY due to glucokinase mutations (GCK-MODY, n = 23), type 1 diabetes (n = 29), latent autoimmune diabetes in adults (LADA, n = 42), and type 2 diabetes (n = 206). Receiver operating characteristic curve analysis was performed to assess discriminative accuracy of 1,5AG for diabetes etiology.Mean (SD range) 1,5AG levels were: GCK-MODY 13.06 microg/ml (5.74-29.74), HNF1A-MODY 4.23 microg/ml (2.12-8.44), type 1 diabetes 3.09 microg/ml (1.45-6.57), LADA 3.46 microg/ml (1.42-8.45), and type 2 diabetes 5.43 (2.12-13.23). Levels in GCK-MODY were higher than in other groups (P < 10(-4) vs. each group). HNF1A-MODY subjects showed no difference in unadjusted 1,5AG levels from type 2 diabetes, type 1 diabetes, and LADA. Adjusting for A1C revealed a difference between HNF1A-MODY and type 2 diabetes (P = 0.001). The discriminative accuracy of unadjusted 1,5AG levels was 0.79 for GCK-MODY versus type 2 diabetes and 0.86 for GCK-MODY versus HNF1A-MODY but was only 0.60 for HNF1A-MODY versus type 2 diabetes.In our dataset, serum 1,5AG performed well in discriminating GCK-MODY from other diabetes subtypes, particularly HNF1A-MODY. Measurement of 1,5AG levels could inform decisions regarding MODY diagnostic testing.

    View details for DOI 10.2337/dc09-1246

    View details for Web of Science ID 000275143700007

    View details for PubMedID 19933992

    View details for PubMedCentralID PMC2809258

  • Assessment of high-sensitivity C-reactive protein levels as diagnostic discriminator of maturity-onset diabetes of the young due to HNF1A mutations. Diabetes care Owen, K. R., Thanabalasingham, G. n., James, T. J., Karpe, F. n., Farmer, A. J., McCarthy, M. I., Gloyn, A. L. 2010; 33 (9): 1919–24

    Abstract

    Despite the clinical importance of an accurate diagnosis in individuals with monogenic forms of diabetes, restricted access to genetic testing leaves many patients with undiagnosed diabetes. Recently, common variation near the HNF1 homeobox A (HNF1A) gene was shown to influence C-reactive protein levels in healthy adults. We hypothesized that serum levels of high-sensitivity C-reactive protein (hs-CRP) could represent a clinically useful biomarker for the identification of HNF1A mutations causing maturity-onset diabetes of the young (MODY).Serum hs-CRP was measured in subjects with HNF1A-MODY (n = 31), autoimmune diabetes (n = 316), type 2 diabetes (n = 240), and glucokinase (GCK) MODY (n = 24) and in nondiabetic individuals (n = 198). The discriminative accuracy of hs-CRP was evaluated through receiver operating characteristic (ROC) curve analysis, and performance was compared with standard diagnostic criteria. Our primary analyses excluded approximately 11% of subjects in whom the single available hs-CRP measurement was >10 mg/l.Geometric mean (SD range) hs-CRP levels were significantly lower (P

    View details for DOI 10.2337/dc10-0288

    View details for PubMedID 20724646

    View details for PubMedCentralID PMC2928334

  • Naturally Occurring Glucokinase Mutations Are Associated with Defects in Posttranslational S-Nitrosylation MOLECULAR ENDOCRINOLOGY Ding, S., Tribble, N. D., Kraft, C. A., Markwardt, M., Gloyn, A. L., Rizzo, M. A. 2010; 24 (1): 171–77

    Abstract

    Posttranslational activation of glucokinase (GCK) through S-nitrosylation has been recently observed in the insulin-secreting pancreatic beta-cell; however, the function of this molecular mechanism in regulating the physiology of insulin secretion is not well understood. To more fully understand the function of posttranslational regulation of GCK, we examined two naturally occurring GCK mutations that map to residues proximal to the S-nitrosylated cysteine and cause mild fasting hyperglycemia (maturity-onset diabetes of the young; subtype glucokinase). The kinetics of recombinantly generated GCK-R369P and GCK-V367M were assessed in vitro. The GCK-R369P protein has greatly reduced catalytic activity (relative activity index 0.05 vs. 1.00 for wild type), whereas the GCK-V367M has near normal kinetics (relative activity index 1.26 vs. 1.00 for wild type). Quantitative imaging and biochemical assays were used to assess the effect of these mutants on the metabolic response to glucose, GCK activation, and S-nitrosylation of GCK in betaTC3 insulinoma cells. Expression of either mutant in betaTC3 cells did not affect the metabolic response to 5 mM glucose. However, expression of either mutant blocked the effects of insulin on glucose-stimulated nicotinamide adenine dinucleotide and nicotinamide adenine dinucleotide phosphate reduction, suggesting defects in posttranslational regulation of GCK. Each of these mutations blocked GCK activation, and prevented posttranslational cysteine S-nitrosylation. Our findings link defects in hormone-regulated GCK S-nitrosylation to hyperglycemia and support a role for posttranslational regulation of GCK S-nitrosylation as a vital regulatory mechanism for glucose-stimulated insulin secretion.

    View details for DOI 10.1210/me.2009-0138

    View details for Web of Science ID 000273071800014

    View details for PubMedID 19934346

    View details for PubMedCentralID PMC2802892

  • Species-Specific Differences in the Expression of the HNF1A, HNF1B and HNF4A Genes PLOS ONE Harries, L. W., Brown, J. E., Gloyn, A. L. 2009; 4 (11): e7855

    Abstract

    The HNF1A, HNF1B and HNF4A genes are part of an autoregulatory network in mammalian pancreas, liver, kidney and gut. The layout of this network appears to be similar in rodents and humans, but inactivation of HNF1A, HNF1B or HNF4A genes in animal models cause divergent phenotypes to those seen in man. We hypothesised that some differences may arise from variation in the expression profile of alternatively processed isoforms between species.We measured the expression of the major isoforms of the HNF1A, HNF1B and HNF4A genes in human and rodent pancreas, islet, liver and kidney by isoform-specific quantitative real-time PCR and compared their expression by the comparative Ct (DeltaDeltaCt) method. We found major changes in the expression profiles of the HNF genes between humans and rodents. The principal difference lies in the expression of the HNF1A gene, which exists as three isoforms in man, but as a single isoform only in rodents. More subtle changes were to the balance of HNF1B and HNF4A isoforms between species; the repressor isoform HNF1B(C) comprised only 6% in human islets compared with 24-26% in rodents (p = 0.006) whereas HNF4A9 comprised 22% of HNF4A expression in human pancreas but only 11% in rodents (p = 0.001).The differences we note in the isoform-specific expression of the human and rodent HNF1A, HNF1B and HNF4A genes may impact on the absolute activity of these genes, and therefore on the activity of the pancreatic transcription factor network as a whole. We conclude that alterations to expression of HNF isoforms may underlie some of the phenotypic variation caused by mutations in these genes.

    View details for DOI 10.1371/journal.pone.0007855

    View details for Web of Science ID 000271851000012

    View details for PubMedID 19924231

    View details for PubMedCentralID PMC2773013

  • RD Lawrence Lecture 2009 Old genes, new tricks: learning about blood glucose regulation from naturally occurring genetic variation in humans DIABETIC MEDICINE Gloyn, A. L. 2009; 26 (11): 1083–89

    Abstract

    The study of rare monogenic forms of diabetes and pancreatic B-cell dysfunction provides an unrivalled opportunity to link a specific change in gene function with precise cellular consequences and clinical phenotype in humans. Over the past 20 years there has been considerable success in determining the genetic aetiology of a number of rare monogenic forms of diabetes, which has had a significant impact on both our understanding of normal physiology and on translational medicine. The impact of these discoveries has been substantial, with insights into both developmental biology and normal physiology. There are clear examples where determining the genetic aetiology for individuals with rare monogenic subtypes of diabetes has led to improved treatment. Although formerly in the shadow of the monogenic diabetes field, over the past 3 years there has been staggering progress in our understanding of the genetic basis of Type 2 diabetes. This has been largely as a result of genome-wide association studies and has seen the list of 'diabetes susceptibility genes' increase from three to close to 20. There is now encouraging evidence to support a potential role for genetics in determining the response of individuals with Type 2 diabetes to different therapeutic options. One of the challenges that lies ahead is determining how the non-coding genetic variants exert their pathogenicity. It is possible that parallels can be drawn from functional work on rare regulatory mutations causing monogenic forms of diabetes. However, it is more likely that comprehensive approaches will be necessary.

    View details for DOI 10.1111/j.1464-5491.2009.02860.x

    View details for Web of Science ID 000271493900002

    View details for PubMedID 19929985

  • Update on Mutations in Glucokinase (GCK), Which Cause Maturity-Onset Diabetes of the Young, Permanent Neonatal Diabetes, and Hyperinsulinemic Hypoglycemia HUMAN MUTATION Osbak, K. K., Colclough, K., Saint-Martin, C., Beer, N. L., Bellanne-Chantelot, C., Ellard, S., Gloyn, A. L. 2009; 30 (11): 1512–26

    Abstract

    Glucokinase is a key regulatory enzyme in the pancreatic beta-cell. It plays a crucial role in the regulation of insulin secretion and has been termed the glucose sensor in pancreatic beta-cells. Given its central role in the regulation of insulin release it is understandable that mutations in the gene encoding glucokinase (GCK) can cause both hyper- and hypoglycemia. Heterozygous inactivating mutations in GCK cause maturity-onset diabetes of the young (MODY) subtype glucokinase (GCK), characterized by mild fasting hyperglycemia, which is present at birth but often only detected later in life during screening for other purposes. Homozygous inactivating GCK mutations result in a more severe phenotype presenting at birth as permanent neonatal diabetes mellitus (PNDM). A growing number of heterozygous activating GCK mutations that cause hypoglycemia have also been reported. A total of 620 mutations in the GCK gene have been described in a total of 1,441 families. There are no common mutations, and the mutations are distributed throughout the gene. The majority of activating mutations cluster in a discrete region of the protein termed the allosteric activator site. The identification of a GCK mutation in patients with both hyper- and hypoglycemia has implications for the clinical course and clinical management of their disorder.

    View details for DOI 10.1002/humu.21110

    View details for Web of Science ID 000271576600003

    View details for PubMedID 19790256

  • Coexpression of the Type 2 Diabetes Susceptibility Gene Variants KCNJ11 E23K and ABCC8 S1369A Alter the ATP and Sulfonylurea Sensitivities of the ATP-Sensitive K+ Channel DIABETES Hamming, K. C., Soliman, D., Matemisz, L. C., Niazi, O., Lang, Y., Gloyn, A. L., Light, P. E. 2009; 58 (10): 2419–24

    Abstract

    In the pancreatic beta-cell, ATP-sensitive K(+) (K(ATP)) channels couple metabolism with excitability and consist of Kir6.2 and SUR1 subunits encoded by KCNJ11 and ABCC8, respectively. Sulfonylureas, which inhibit the K(ATP) channel, are used to treat type 2 diabetes. Rare activating mutations cause neonatal diabetes, whereas the common variants, E23K in KCNJ11 and S1369A in ABCC8, are in strong linkage disequilibrium, constituting a haplotype that predisposes to type 2 diabetes. To date it has not been possible to establish which of these represents the etiological variant, and functional studies are inconsistent. Furthermore, there have been no studies of the S1369A variant or the combined effect of the two on K(ATP) channel function.The patch-clamp technique was used to study the nucleotide sensitivity and sulfonylurea inhibition of recombinant human K(ATP) channels containing either the K23/A1369 or E23/S1369 variants.ATP sensitivity of the K(ATP) channel was decreased in the K23/A1369 variant (half-maximal inhibitory concentration [IC(50)] = 8.0 vs. 2.5 mumol/l for the E23/S1369 variant), although there was no difference in ADP sensitivity. The K23/A1369 variant also displayed increased inhibition by gliclazide, an A-site sulfonylurea drug (IC(50) = 52.7 vs. 188.7 nmol/l for the E23/S1369 variant), but not by glibenclamide (AB site) or repaglinide (B site).Our findings indicate that the common K23/A1369 variant K(ATP) channel displays decreased ATP inhibition that may contribute to the observed increased risk for type 2 diabetes. Moreover, the increased sensitivity of the K23/A1369 variant to the A-site sulfonylurea drug gliclazide may provide a pharmacogenomic therapeutic approach for patients with type 2 diabetes who are homozygous for both risk alleles.

    View details for DOI 10.2337/db09-0143

    View details for Web of Science ID 000270776200031

    View details for PubMedID 19587354

    View details for PubMedCentralID PMC2750221

  • Low Frequency Variants in the Exons Only Encoding Isoform A of HNF1A Do Not Contribute to Susceptibility to Type 2 Diabetes PLOS ONE Jafar-Mohammadi, B., Groves, C. J., Owen, K. R., Frayling, T. M., Hattersley, A. T., McCarthy, M. I., Gloyn, A. L. 2009; 4 (8): e6615

    Abstract

    There is considerable interest in the hypothesis that low frequency, intermediate penetrance variants contribute to the proportion of Type 2 Diabetes (T2D) susceptibility not attributable to the common variants uncovered through genome-wide association approaches. Genes previously implicated in monogenic and multifactorial forms of diabetes are obvious candidates in this respect. In this study, we focussed on exons 8-10 of the HNF1A gene since rare, penetrant mutations in these exons (which are only transcribed in selected HNF1A isoforms) are associated with a later age of diagnosis of Maturity onset diabetes of the young (MODY) than mutations in exons 1-7. The age of diagnosis in the subgroup of HNF1A-MODY individuals with exon 8-10 mutations overlaps with that of early multifactorial T2D, and we set out to test the hypothesis that these exons might also harbour low-frequency coding variants of intermediate penetrance that contribute to risk of multifactorial T2D.We performed targeted capillary resequencing of HNF1A exons 8-10 in 591 European T2D subjects enriched for genetic aetiology on the basis of an early age of diagnosis (< or =45 years) and/or family history of T2D (> or =1 affected sibling). PCR products were sequenced and compared to the published HNF1A sequence. We identified several variants (rs735396 [IVS9-24T>C], rs1169304 [IVS8+29T>C], c.1768+44C>T [IVS9+44C>T] and rs61953349 [c.1545G>A, p.T515T] but no novel non-synonymous coding variants were detected.We conclude that low frequency, nonsynonymous coding variants in the terminal exons of HNF1A are unlikely to contribute to T2D-susceptibility in European samples. Nevertheless, the rationale for seeking low-frequency causal variants in genes known to contain rare, penetrant mutations remains strong and should motivate efforts to screen other genes in a similar fashion.

    View details for DOI 10.1371/journal.pone.0006615

    View details for Web of Science ID 000268935900024

    View details for PubMedID 19672314

    View details for PubMedCentralID PMC2720540

  • Partial lipodystrophy and insulin resistant diabetes in a patient with a homozygous nonsense mutation in CIDEC EMBO MOLECULAR MEDICINE Rubio-Cabezas, O., Puri, V., Murano, I., Saudek, V., Semple, R. K., Dash, S., Hyden, C. S., Bottomley, W., Vigouroux, C., Magre, J., Raymond-Barker, P., Murgatroyd, P. R., Chawla, A., Skepper, J. N., Chatterjee, V., Suliman, S., Patch, A., Agarwal, A. K., Garg, A., Barroso, I., Cinti, S., Czech, M. P., Argente, J., O'Rahilly, S., Savage, D. B., LD Screening Consortium 2009; 1 (5): 280–87

    Abstract

    Lipodystrophic syndromes are characterized by adipose tissue deficiency. Although rare, they are of considerable interest as they, like obesity, typically lead to ectopic lipid accumulation, dyslipidaemia and insulin resistant diabetes. In this paper we describe a female patient with partial lipodystrophy (affecting limb, femorogluteal and subcutaneous abdominal fat), white adipocytes with multiloculated lipid droplets and insulin-resistant diabetes, who was found to be homozygous for a premature truncation mutation in the lipid droplet protein cell death-inducing Dffa-like effector C (CIDEC) (E186X). The truncation disrupts the highly conserved CIDE-C domain and the mutant protein is mistargeted and fails to increase the lipid droplet size in transfected cells. In mice, Cidec deficiency also reduces fat mass and induces the formation of white adipocytes with multilocular lipid droplets, but in contrast to our patient, Cidec null mice are protected against diet-induced obesity and insulin resistance. In addition to describing a novel autosomal recessive form of familial partial lipodystrophy, these observations also suggest that CIDEC is required for unilocular lipid droplet formation and optimal energy storage in human fat.

    View details for DOI 10.1002/emmm.200900037

    View details for Web of Science ID 000273563300005

    View details for PubMedID 20049731

    View details for PubMedCentralID PMC2891108

  • Type 2 Diabetes Susceptibility Gene TCF7L2 and Its Role in beta-Cell Function DIABETES Gloyn, A. L., Braun, M., Rorsman, P. 2009; 58 (4): 800–802

    View details for DOI 10.2337/db09-0099

    View details for Web of Science ID 000264819700004

    View details for PubMedID 19336690

    View details for PubMedCentralID PMC2661580

  • Update of Mutations in the Genes Encoding the Pancreatic Beta-Cell K-ATP Channel Subunits Kir6.2 (KCNJ11) and Sulfonylurea Receptor 1 (ABCC8) in Diabetes Mellitus and Hyperinsulinism HUMAN MUTATION Flanagan, S. E., Clauin, S., Bellanne-Chantelot, C., de Lonlay, P., Harries, L. W., Gloyn, A. L., Ellard, S. 2009; 30 (2): 170–80

    Abstract

    The beta-cell ATP-sensitive potassium (K(ATP)) channel is a key component of stimulus-secretion coupling in the pancreatic beta-cell. The channel couples metabolism to membrane electrical events bringing about insulin secretion. Given the critical role of this channel in glucose homeostasis it is therefore not surprising that mutations in the genes encoding for the two essential subunits of the channel can result in both hypo- and hyperglycemia. The channel consists of four subunits of the inwardly rectifying potassium channel Kir6.2 and four subunits of the sulfonylurea receptor 1 (SUR1). It has been known for some time that loss of function mutations in KCNJ11, which encodes for Kir6.2, and ABCC8, which encodes for SUR1, can cause oversecretion of insulin and result in hyperinsulinism of infancy, while activating mutations in KCNJ11 and ABCC8 have recently been described that result in the opposite phenotype of diabetes. This review focuses on reported mutations in both genes, the spectrum of phenotypes, and the implications for treatment on diagnosing patients with mutations in these genes.

    View details for DOI 10.1002/humu.20838

    View details for Web of Science ID 000263254200006

    View details for PubMedID 18767144

  • Severe insulin resistance and intrauterine growth deficiency associated with haploinsufficiency for INSR and CHN2: new insights into synergistic pathways involved in growth and metabolism. Diabetes Suliman, S. G., Stanik, J. n., McCulloch, L. J., Wilson, N. n., Edghill, E. L., Misovicova, N. n., Gasperikova, D. n., Sandrikova, V. n., Elliott, K. S., Barak, L. n., Ellard, S. n., Volpi, E. V., Klimes, I. n., Gloyn, A. L. 2009; 58 (12): 2954–61

    Abstract

    Digenic causes of human disease are rarely reported. Insulin via its receptor, which is encoded by INSR, plays a key role in both metabolic and growth signaling pathways. Heterozygous INSR mutations are the most common cause of monogenic insulin resistance. However, growth retardation is only reported with homozygous or compound heterozygous mutations. We describe a novel translocation [t(7,19)(p15.2;p13.2)] cosegregating with insulin resistance and pre- and postnatal growth deficiency. Chromosome translocations present a unique opportunity to identify modifying loci; therefore, our objective was to determine the mutational mechanism resulting in this complex phenotype.Breakpoint mapping was performed by fluorescence in situ hybridization (FISH) on patient chromosomes. Sequencing and gene expression studies of disrupted and adjacent genes were performed on patient-derived tissues. RESULTS Affected individuals had increased insulin, C-peptide, insulin-to-C-peptide ratio, and adiponectin levels consistent with an insulin receptoropathy. FISH mapping established that the translocation breakpoints disrupt INSR on chromosome 19p15.2 and CHN2 on chromosome 7p13.2. Sequencing demonstrated INSR haploinsufficiency accounting for elevated insulin levels and dysglycemia. CHN2 encoding beta-2 chimerin was shown to be expressed in insulin-sensitive tissues, and its disruption was shown to result in decreased gene expression in patient-derived adipose tissue.We present a likely digenic cause of insulin resistance and growth deficiency resulting from the combined heterozygous disruption of INSR and CHN2, implicating CHN2 for the first time as a key element of proximal insulin signaling in vivo.

    View details for DOI 10.2337/db09-0787

    View details for PubMedID 19720790

    View details for PubMedCentralID PMC2780873

  • Mutations in the third gene shown to alter fasting glucose levels in the population (G6PC2) are not a common cause of monogenic forms of pancreatic B-cell dysfunction DIABETIC MEDICINE Edghill, E. L., McCulloch, L., Fulton, P., Beer, N., Hattersley, A. T., Gloyn, A. L. 2009; 26 (1): 113–14
  • Prevalence of GCK mutations in individuals screened for fasting hyperglycaemia DIABETOLOGIA Gloyn, A. L., van de Bunt, M., Stratton, I. M., Lonie, L., Tucker, L., Ellard, S., Holman, R. R. 2009; 52 (1): 172–74

    View details for DOI 10.1007/s00125-008-1188-4

    View details for Web of Science ID 000261375400024

    View details for PubMedID 19002431

  • The P446L variant in GCKR associated with fasting plasma glucose and triglyceride levels exerts its effect through increased glucokinase activity in liver. Human molecular genetics Beer, N. L., Tribble, N. D., McCulloch, L. J., Roos, C. n., Johnson, P. R., Orho-Melander, M. n., Gloyn, A. L. 2009; 18 (21): 4081–88

    Abstract

    Genome-wide association studies have identified a number of signals for both Type 2 Diabetes and related quantitative traits. For the majority of loci, the transition from association signal to mutational mechanism has been difficult to establish. Glucokinase (GCK) regulates glucose storage and disposal in the liver where its activity is regulated by glucokinase regulatory protein (GKRP; gene name GCKR). Fructose-6 and fructose-1 phosphate (F6P and F1P) enhance or reduce GKRP-mediated inhibition, respectively. A common GCKR variant (P446L) is reproducibly associated with triglyceride and fasting plasma glucose levels in the general population. The aim of this study was to determine the mutational mechanism responsible for this genetic association. Recombinant human GCK and both human wild-type (WT) and P446L-GKRP proteins were generated. GCK kinetic activity was observed spectrophotometrically using an NADP(+)-coupled assay. WT and P446L-GKRP-mediated inhibition of GCK activity and subsequent regulation by phosphate esters were determined. Assays matched for GKRP activity demonstrated no difference in dose-dependent inhibition of GCK activity or F1P-mediated regulation. However, the response to physiologically relevant F6P levels was significantly attenuated with P446L-GKRP (n = 18; P

    View details for DOI 10.1093/hmg/ddp357

    View details for PubMedID 19643913

    View details for PubMedCentralID PMC2758140

  • Identification of a novel beta-cell glucokinase (GCK) promoter mutation (-71G>C) that modulates GCK gene expression through loss of allele-specific Sp1 binding causing mild fasting hyperglycemia in humans. Diabetes Gasperíková, D. n., Tribble, N. D., Staník, J. n., Hucková, M. n., Misovicová, N. n., van de Bunt, M. n., Valentínová, L. n., Barrow, B. A., Barák, L. n., Dobránsky, R. n., Bereczková, E. n., Michálek, J. n., Wicks, K. n., Colclough, K. n., Knight, J. C., Ellard, S. n., Klimes, I. n., Gloyn, A. L. 2009; 58 (8): 1929–35

    Abstract

    Inactivating mutations in glucokinase (GCK) cause mild fasting hyperglycemia. Identification of a GCK mutation has implications for treatment and prognosis; therefore, it is important to identify these individuals. A significant number of patients have a phenotype suggesting a defect in glucokinase but no abnormality of GCK. We hypothesized that the GCK beta-cell promoter region, which currently is not routinely screened, could contain pathogenic mutations; therefore, we sequenced this region in 60 such probands.The beta-cell GCK promoter was sequenced in patient DNA. The effect of the identified novel mutation on GCK promoter activity was assessed using a luciferase reporter gene expression system. Electrophoretic mobility shift assays (EMSAs) were used to determine the impact of the mutation on Sp1 binding.A novel -71G>C mutation was identified in a nonconserved region of the human promoter sequence in six apparently unrelated probands. Family testing established cosegregation with fasting hyperglycemia (> or = 5.5 mmol/l) in 39 affected individuals. Haplotype analysis in the U.K. family and four of the Slovakian families demonstrated that the mutation had arisen independently. The mutation maps to a potential transcriptional activator binding site for Sp1. Reporter assays demonstrated that the mutation reduces promoter activity by up to fourfold. EMSAs demonstrated a dramatic reduction in Sp1 binding to the promoter sequence corresponding to the mutant allele.A novel beta-cell GCK promoter mutation was identified that significantly reduces gene expression in vitro through loss of regulation by Sp1. To ensure correct diagnosis of potential GCK-MODY (maturity-onset diabetes of the young) cases, analysis of the beta-cell GCK promoter should be included.

    View details for DOI 10.2337/db09-0070

    View details for PubMedID 19411616

    View details for PubMedCentralID PMC2712784

  • Genetics: how the UKPDS contributed to determining the genetic landscape of Type 2 diabetes DIABETIC MEDICINE Gloyn, A. L., McCarthy, M. I. 2008; 25: 35–40

    Abstract

    The identification and functional characterisation of genetic variants that either cause or predispose to diabetes is a major focus of biomedical research. The molecular basis is now known for the majority of monogenic forms of diabetes arising from pancreatic beta-cell dysfunction; however finding the genetic variants underlying susceptibility to Type 2 diabetes (T2DM) has been a greater technical, statistical and biological challenge. The advent of biology-agnostic approaches made possible by the improved arsenal of research platforms and genetic tools available has increased the number of known T2DM genes dramatically and provided important insights into the pathophysiology of T2DM. Over the past 18 months, the list of T2DM susceptibility genes has grown from three to close to 20, illustrating the substantial progress which has been made. These recent milestones have not only illustrated the limited knowledge we have of the pancreatic beta-cell, but have also reinforced our belief in the involvement of common genetic variants in the genes involved in monogenic forms of diabetes in the susceptibility to T2DM and have clearly shown a primary role for pancreatic beta-cell dysfunction in T2DM. Both of these concepts were explored in the early work of the UK Prospective Diabetes Study (UKPDS) genetics research groups.

    View details for DOI 10.1111/j.1464-5491.2008.02500.x

    View details for Web of Science ID 000259037200008

    View details for PubMedID 18717977

  • Activating glucokinase (GCK) Mutations as a cause of medically responsive congenital hyperinsulinism: prevalence in children and characterisation of a novel GCK mutation EUROPEAN JOURNAL OF ENDOCRINOLOGY Christesen, H. T., Tribble, N. D., Molven, A., Siddiqui, J., Sandal, T., Brusgaard, K., Ellard, S., Njolstad, P. R., Alm, J., Jacobsen, B., Hussain, K., Gloyn, A. L. 2008; 159 (1): 27–34

    Abstract

    Activating glucokinase (GCK) mutations are a rarely reported cause of congenital hyperinsulinism (CHI), but the prevalence of GCK mutations is not known.From a pooled cohort of 201 non-syndromic children with CHI from three European referral centres (Denmark, n=141; Norway, n=26; UK, n=34), 108 children had no K(ATP)-channel (ABCC8/KCNJ11) gene abnormalities and were screened for GCK mutations. Novel GCK mutations were kinetically characterised.In five patients, four heterozygous GCK mutations (S64Y, T65I, W99R and A456V) were identified, out of which S64Y was novel. Two of the mutations arose de novo, three were dominantly inherited. All the five patients were medically responsive. In the combined Danish and Norwegian cohort, the prevalence of GCK-CHI was estimated to be 1.2% (2/167, 95% confidence interval (CI) 0-2.8%) of all the CHI patients. In the three centre combined cohort of 72 medically responsive children without K(ATP)-channel mutations, the prevalence estimate was 6.9% (5/72, 95% CI 1.1-12.8%). All activating GCK mutations mapped to the allosteric activator site. The novel S64Y mutation resulted in an increased affinity for the substrate glucose (S(0.5) 1.49+/-0.08 and 7.39+/-0.05 mmol/l in mutant and wild-type proteins respectively), extrapolating to a relative activity index of approximately 22 compared with the wild type.In the largest study performed to date on GCK in children with CHI, GCK mutations were found only in medically responsive children who were negative for ABCC8 and KCNJ11 mutations. The estimated prevalence (approximately 7%) suggests that screening for activating GCK mutations is warranted in those patients.

    View details for DOI 10.1530/EJE-08-0203

    View details for Web of Science ID 000257855800005

    View details for PubMedID 18450771

  • Permanent neonatal diabetes mellitus caused by a novel homozygous (T168A) glucokinase (GCK) mutation: Initial response to oral sulphonylurea therapy JOURNAL OF PEDIATRICS Turkkahraman, D., Brcan, I., Tribble, N. D., Akcurin, S., Ellard, S., Gloyn, A. L. 2008; 153 (1): 122–26

    Abstract

    To evaluate the clinical response to sulphonylurea treatment in a child with a homozygous T168A GCK (glucokinase) mutation, causing permanent neonatal diabetes mellitus (PNDM).Oral glibenclamide was given for 3 months. Pancreatic beta cell function was assessed by a glucagon stimulation test. Mutant and wild-type (WT) GCK were characterized.Sulphonylurea treatment resulted in a 12-fold increase in basal and stimulated C-peptide levels. HbA1c levels were reduced from 9.4% to 8.1% on a reduced insulin dose (0.85 to 0.60 U/kg/day). Mutant T168A-GST-GCK showed reduced kinetic activity (0.02 fold) compared to WT.Sulphonylureas can close the adenosine triphosphate (ATP)-sensitive potassium channel and elicit insulin secretion, but the ATP generated from metabolism is insufficient to fully restore insulin secretory capacity. Nonetheless, sulphonylurea treatment should be tried in patients with GCK-PNDM, particularly those with mutations resulting in less severe kinetic defects, in whom improved glycemic control may be obtained with lower doses of insulin.

    View details for DOI 10.1016/j.jpeds.2007.12.037

    View details for Web of Science ID 000257154800029

    View details for PubMedID 18571549

  • Glucokinase (GCK) and other susceptibility genes for beta-cell dysfunction: the candidate approach Gloyn, A. L., Tribble, N. D., van de Bunt, M., Barrett, A., Johnson, P. V. PORTLAND PRESS LTD. 2008: 306–11

    Abstract

    There are well-documented examples in the literature of where determining the genetic aetiology of a disorder has provided insights into important regulatory pathways and protein interactions, and, more recently, has led to improved treatment options for patients. The studies of monogenic forms of beta-cell dysfunction are no exception. Naturally occurring mutations in the gene for the beta-cell enzyme glucokinase (GCK) result in both hyper- and hypo-glycaemia. Over 200 mutations have been described, and careful study of the mutational mechanisms for a number of these has provided important insights into glucokinase regulation. Increased understanding of post-translational regulatory mechanisms holds the promise of novel pharmacotherapeutic options for the treatment of T2DM (Type 2 diabetes mellitus). It is well established that common genetic variation in genes involved in monogenic forms of beta-cell dysfunction contributes to susceptibility to T2DM. Recent genome-wide scans for association have identified a number of novel T2DM susceptibility genes which probably influence beta-cell mass and/or function. Their identification allows the investigation of the role of rare mutations in monogenic beta-cell dysfunction. Current results indicate the importance of these genes in pancreatic development and suggest that mutations which result in a severe functional defect could be lethal.

    View details for DOI 10.1042/BST0360306

    View details for Web of Science ID 000256609000009

    View details for PubMedID 18481947

  • Gene duplications resulting in over expression of glucokinase are not a common cause of hypoglycaemia of infancy in humans MOLECULAR GENETICS AND METABOLISM van de Bunt, M., Edghill, E. L., Hussain, K., Ellard, S., Gloyn, A. L. 2008; 94 (2): 268–69

    View details for DOI 10.1016/j.ymgme.2008.01.008

    View details for Web of Science ID 000256371900020

    View details for PubMedID 18325809

    View details for PubMedCentralID PMC2427397

  • Prevalence and clinical characteristics of maternally inherited diabetes and deafness caused by the mt3243A > G mutation in young adult diabetic subjects in Sri Lanka DIABETIC MEDICINE Katulanda, P., Groves, C. J., Barrett, A., Sheriff, R., Matthews, D. R., McCarthy, M. I., Gloyn, A. L. 2008; 25 (3): 370–74

    Abstract

    The maternally inherited mt3243A > G mutation is associated with a variable clinical phenotype including diabetes and deafness (MIDD). We aimed to determine the prevalence and clinical characteristics of MIDD in a large South Asian cohort of young adult-onset diabetic patients from Sri Lanka.DNA was available from 994 subjects (age of diagnosis 16-40 years, age at recruitment < or = 45 years). Mutation screening was performed using a QRT-PCR method on an ABI 7900HT system using sequence-specific probes. Samples with heteroplasm > or = 5.0% were considered positive.Nine (four males) mutation-positive subjects were identified (prevalence 0.9%). They were diagnosed at a younger age (25.9 +/- 4.8 years vs. 31.9 +/- 5.6 years, P = 0.002) and were lean (body mass index [BMI] 18.7 +/- 2.7 kg/m(2) vs. 24.7 +/- 4.0 kg/m(2), P < 0.001) compared to NMCs. One mutation-positive subject (11.1%) had metabolic syndrome, compared to 633 (64.3%) of NMCs. Insulin therapy within 6 months of diagnosis was used in four (44.0%) carriers compared to 6.9% of NMCs (P = 0.002). Combined screening criteria of any two of maternal history of diabetes, personal history of hearing impairment and family history of hearing impairment only identified five (55%) of the carriers, with a positive predictive value of 7.4%.The prevalence of mt3243A > G mutation among young adult-onset diabetic subjects from Sri Lanka was 0.9%. Our study demonstrates that a maternal family history of diabetes and either a personal and/or family history of deafness only distinguish half of patients with MIDD from Sri Lankan subjects with young-onset diabetes.

    View details for DOI 10.1111/j.1464-5491.2007.02377.x

    View details for Web of Science ID 000253609300020

    View details for PubMedID 18279408

  • Maturity onset diabetes of the young without gene mutation (MODYX) is rare in children with autosomal dominant diabetes in the UK Porter, J. R., Ellardw, S., Hattersleyw, A., Gloyn, A., Clarky, P., Barrett, T. G. WILEY. 2008
  • Permanent neonatal diabetes mellitus caused by a novel homozygous (T168A) glucokinase (GCK) mutation: response to oral sulphonylurea therapy Gloyn, A. L., Bircanw, I., Tribble, N. D., Akcurinw, S., Ellard, S., Turkkahramanw, D. WILEY. 2008
  • Congenital Hyperinsullnism: Prevalence Prevalence of activating glucokinase mutations and a novel mutation Christesen, H., Tribble, N. D., Molven, A., Siddiqi, J., Sandal, T., Brusgaard, K., Ellard, S., Njolstad, P. R., Alm, J., Jacobsen, B., Hussain, K., Gloyn, A. L. KARGER. 2008: 41
  • Heterogeneity in disease severity in a family with a novel G68V GCK activating mutation causing persistent hyperinsulinaemic hypoglycaemia of infancy DIABETIC MEDICINE Wabitsch, M., Lahr, G., Van de Bunt, M., Marchant, C., Lindnert, M., von Puttkamer, J., Fenneberg, A., Debatin, K. M., Klein, R., Ellard, S., Clark, A., Gloyn, A. L. 2007; 24 (12): 1393–99

    Abstract

    Glucokinase (GCK)-activating mutations cause persistent hyperinsulinaemic hypoglycaemia of infancy (PHHI). GCK-PHHI patients have regulated insulin secretion and can usually be treated with diazoxide. The six reported cases suggest that the severity of the mutation predicts the clinical phenotype. The aim of this study was to relate genotype to phenotype [clinical phenotype, glucose-stimulated insulin release (GSIR) and GCK functional analysis] in a large pedigree with eight affected individuals.The genes encoding B-cell GCK and the K(ATP) channel subunits (ABCC8 and KCNJ11) were sequenced to identify mutations for functional analysis. Genetic variants influencing B-cell function were genotyped in affected individuals. Islet secretory capacity was determined by oral glucose tolerance testA novel GCK mutation (G68V) co-segregating with hypoglycaemia was identified in eight family members. Kinetic analysis revealed that G68V-GCK activity is ~16 times more than wild-type-GCK with an increased affinity for glucose [concentration at half maximal activation (S(0.5)) 1.94 +/- 0.16 vs. 7.43 +/- 0.12, mutant vs. wild type, mean +/- sem]. Mathematical modelling predicted a threshold for GSIR of 1.9 mmol/l in the mutant. Oral glucose tolerance tests showed regulated insulin secretion. The severity of hypoglycaemia and related symptoms in affected subjects were heterogeneous. Clinical presentations were asymptomatic (n = 1), extreme hunger (n = 3), seizures (n = 2) and loss of consciousness (n = 2); 7/8 were managed with diet but the proband was treated with diazoxide and octreotide. Phenotypic modification by a second mutation in the K(ATP) channel genes (ABCC8, KCNJ11) or by common genetic variants in KCNJ11, GCK and TCF7L2 was excluded.The novel activating GCK mutation G68V is associated with variable phenotypic severity, supporting modification of GSIR by genetic and/or environmental factors.

    View details for DOI 10.1111/j.1464-5491.2007.02285.x

    View details for Web of Science ID 000251250200013

    View details for PubMedID 17976205

  • Mutations in HHEX are not a common cause of monogenic forms of beta cell dysfunction DIABETOLOGIA Minton, J. L., van de Bunt, M., Boustred, C., Hussain, K., Hattersley, A. T., Ellard, S., Gloyn, A. L. 2007; 50 (9): 2019–22

    View details for DOI 10.1007/s00125-007-0748-3

    View details for Web of Science ID 000248771800027

    View details for PubMedID 17618412

  • Monogenic disorders of the pancreatic beta-cell: personalizing treatment for rare forms of diabetes and hypoglycemia. Personalized medicine van de Bunt, M., Gloyn, A. L. 2007; 4 (3): 247–59

    Abstract

    Over the past 10-20years, our understanding of the genetic etiology of monogenic disorders of the pancreatic beta-cell has greatly improved. This has enabled clinicians to provide patients with more accurate information regarding prognosis and inheritance and has influenced treatment. Maturity-onset diabetes of the young and neonatal diabetes are two such examples. Patients with maturity-onset diabetes of the young due to glucokinase mutations can usually be managed by dietalone, while those affected by HNF-1alpha and HNF-4alpha mutations respond well to low doses of sulfonylureas. The identification of mutations in the ATP-dependent potassium channel genes KCNJ11 and ABCC8 as the most common cause of permanent neonatal diabetes has improved treatment regimes for affected children. In addition to enabling patients to stop insulin injections, their glycemic control has also improved. These advances show the importance of unravelling the genetics of a disease to achieve the best individualized treatment for the patients affected.

    View details for DOI 10.2217/17410541.4.3.247

    View details for PubMedID 29788672

  • Cell biology assessment of glucokinase mutations V62M and G72R in pancreatic beta-cells - Evidence for cellular instability of catalytic activity DIABETES Arden, C., Trainer, A., de la Iglesia, N., Scougall, K. T., Gloyn, A. L., Lange, A. J., Shaw, J. M., Matschinsky, F. M., Agius, L. 2007; 56 (7): 1773–82

    Abstract

    Mutations in the glucokinase (GK) gene cause defects in blood glucose homeostasis. In some cases (V62M and G72R), the phenotype cannot be explained by altered enzyme kinetics or protein instability. We used transient and stable expression of green fluorescent protein (GFP) GK chimaeras in MIN6 beta-cells to study the phenotype defect of V62M and G72R. GK activity in lysates of MIN6 cell lines stably expressing wild-type or mutant GFP GK showed the expected affinity for glucose and response to pharmacological activators, indicating the expression of catalytically active enzymes. MIN6 cells stably expressing GFP V62M or GFP G72R had a lower GK activity-to-GK immunoreactivity ratio and GK activity-to-GK mRNA ratio but not GK immunoreactivity-to-GK mRNA ratio than wild-type GFP GK. Heterologous expression of liver 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (PFK2/FDP2) in cell lines increased GK activity for wild-type GK and V62M but not for G72R, whereas expression of liver GK regulatory protein (GKRP) increased GK activity for wild type but not V62M or G72R. Lack of interaction of these mutants with GKRP was also evident in hepatocyte transfections from the lack of nuclear accumulation. These results suggest that cellular loss of GK catalytic activity rather than impaired translation or enhanced protein degradation may account for the hyperglycemia in subjects with V62M and G72R mutations.

    View details for DOI 10.2337/db06-1151

    View details for Web of Science ID 000247768000003

    View details for PubMedID 17389332

  • Mutations in ATP-sensitive K+ channel genes cause transient neonatal diabetes and permanent diabetes in childhood or adulthood DIABETES Flanagan, S. E., Patch, A., Mackay, D. G., Edghill, E. L., Gloyn, A. L., Robinson, D., Shield, J. H., Temple, K., Ellard, S., Hattersley, A. T. 2007; 56 (7): 1930–37

    Abstract

    Transient neonatal diabetes mellitus (TNDM) is diagnosed in the first 6 months of life, with remission in infancy or early childhood. For approximately 50% of patients, their diabetes will relapse in later life. The majority of cases result from anomalies of the imprinted region on chromosome 6q24, and 14 patients with ATP-sensitive K+ channel (K(ATP) channel) gene mutations have been reported. We determined the 6q24 status in 97 patients with TNDM. In patients in whom no abnormality was identified, the KCNJ11 gene and/or ABCC8 gene, which encode the Kir6.2 and SUR1 subunits of the pancreatic beta-cell K(ATP) channel, were sequenced. K(ATP) channel mutations were found in 25 of 97 (26%) TNDM probands (12 KCNJ11 and 13 ABCC8), while 69 of 97 (71%) had chromosome 6q24 abnormalities. The phenotype associated with KCNJ11 and ABCC8 mutations was similar but markedly different from 6q24 patients who had a lower birth weight and who were diagnosed and remitted earlier (all P < 0.001). K(ATP) channel mutations were identified in 26 additional family members, 17 of whom had diabetes. Of 42 diabetic patients, 91% diagnosed before 6 months remitted, but those diagnosed after 6 months had permanent diabetes (P < 0.0001). K(ATP) channel mutations account for 89% of patients with non-6q24 TNDM and result in a discrete clinical subtype that includes biphasic diabetes that can be treated with sulfonylureas. Remitting neonatal diabetes was observed in two of three mutation carriers, and permanent diabetes occurred after 6 months of age in subjects without an initial diagnosis of neonatal diabetes.

    View details for DOI 10.2337/db07-0043

    View details for Web of Science ID 000247768000022

    View details for PubMedID 17446535

  • Relationship between E23K (an established type II diabetes-susceptibility variant within KCNJ11), polycystic ovary syndrome and androgen levels EUROPEAN JOURNAL OF HUMAN GENETICS Barber, T. M., Bennett, A. J., Gloyn, A. L., Groves, C. J., Sovio, U., Ruokonen, A., Martikainen, H., Pouta, A., Taponen, S., Weedon, M. N., Hartikainen, A., Wass, J. H., Jarvelin, M., Zeggini, E., Franks, S., McCarthy, M. I. 2007; 15 (6): 679–84

    Abstract

    Polycystic ovary syndrome (PCOS) is strongly associated with hyperinsulinaemia and type II diabetes (T2D). Sequence variation within KCNJ11 (encoding Kir6.2, the beta-cell inwardly rectifying potassium channel) is implicated in the pathogenesis of neonatal diabetes, hyperinsulinaemia of infancy and multifactorial T2D. Comprehensive tagging studies have demonstrated that the KCNJ11 E23K variant (or ABCC8 A1369S in LD>0.9) is responsible for the known association between KCNJ11 and T2D. Given the phenotypic overlap between PCOS and T2D, we investigated whether E23K is involved in susceptibility to PCOS and related traits. Case-control analyses for the KCNJ11 E23K variant were performed in (a) 374 PCOS cases and 2574 controls of UK British/Irish origin, and (b) 550 women with PCOS symptoms and 1114 controls from a Finnish birth cohort. The relationship between E23K genotype and androgen levels (a key intermediate phenotype relevant to PCOS) in 1380 samples was studied. The UK case-control analysis revealed no association between E23K genotypes and PCOS status (P=0.49; Cochran-Armitage test), and no significant relationship between E23K genotype and androgen measures in the samples for which these phenotypes were available (P=0.19). Similarly, the Finnish case-control analysis showed no association between E23K genotypes and PCOS status (P=0.75; Cochran-Armitage test), and no significant relationship between E23K genotype and androgen measures in the samples for which these phenotypes were available (Finnish controls, P=0.25; Finnish cases, P=0.08). In conclusion, these data (involving >4600 subjects) provide no evidence that common variants of the KCNJ11 E23K polymorphism have a major influence on PCOS susceptibility, though modest effect sizes (OR<1.25) cannot be excluded.

    View details for DOI 10.1038/sj.ejhg.5201802

    View details for Web of Science ID 000246792100010

    View details for PubMedID 17342155

  • Origin of de novo KCNJ11 mutations and risk of neonatal diabetes for subsequent siblings JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM Edghill, E. L., Gloyn, A. L., Goriely, A., Harries, L. W., Flanagan, S. E., Rankin, J., Hattersley, A. T., Ellard, S. 2007; 92 (5): 1773–77

    Abstract

    Activating mutations in the KCNJ11 gene, which encodes the Kir6.2 subunit of the pancreatic beta-cell K(ATP) channel, result in permanent and transient neonatal diabetes. The majority of KCNJ11 mutations are spontaneous, but the parental origin of these mutations is not known.Our objective was to determine the parental origin of de novo KCNJ11 mutations and investigate the possibility of mosaicism in transmitting parents.We identified 68 index cases with a KCNJ11 mutation where neither parent was known to be affected. DNA was available from both parents of 41 probands. The parental origin of the mutation was determined in 18 families by examination of pedigrees, microsatellite analysis, or allele-specific PCR.A nonsignificant excess of paternally derived mutations was found with 13 of 18 (72%) shown to have arisen on the paternal allele. There was no evidence to suggest an association with increased age at conception. In two families, there were half-siblings with permanent neonatal diabetes born to an unaffected father, suggesting germline mosaicism that was confirmed by the presence of the R201C mutation in one father's semen. Somatic mosaicism was detected in one unaffected mother, and this mutation will also be present in her germ cells.De novo KCNJ11 mutations can arise either during gametogenesis or embryogenesis. The possibility of germline mosaicism means that future siblings are at increased risk of neonatal diabetes, and we recommend that molecular genetic testing is routinely offered at birth for subsequent siblings of children with de novo KCNJ11 mutations.

    View details for DOI 10.1210/jc.2006-2817

    View details for Web of Science ID 000246221200036

    View details for PubMedID 17327377

  • Asian MODY: are we missing an important diagnosis? DIABETIC MEDICINE Porter, J. R., Rangasami, J. J., Ellard, S., Gloyn, A. L., Shields, B. M., Edwards, J., Anderson, J. M., Shaw, N. J., Hattersley, A. T., Frayling, T. M., Plunkett, M., Barrett, T. G. 2006; 23 (11): 1257–60

    Abstract

    Maturity onset diabetes of the young (MODY) is a monogenic form of diabetes where correct diagnosis alters treatment, prognosis and genetic counselling. The first UK survey of childhood MODY identified 20 White, but no Asian children with MODY. We hypothesized that MODY causes diabetes in UK Asians, but is underdiagnosed.Children with dominant family histories of diabetes were recruited. Direct sequencing for mutations in the two most common MODY genes; HNF1A (TCF1) and GCK was performed in autoantibody-negative probands. We also compared MODY testing data for Asian and White cases from the Exeter MODY database, to 2001 UK census data.We recruited 30 families and identified three Asian families with MODY gene mutations (two HNF1A, one GCK) and three White UK families (two HNF1A, one GCK). Heterozygous MODY phenotypes were similar in Asians and Whites. Only eight (0.5%) of 1369 UK referrals for MODY testing were known to be Asian, but in 2001 Asians represented 4% of the English/Welsh population and have a higher prevalence of diabetes.We identified three cases of childhood MODY in UK Asians and demonstrated reduced rates of MODY testing in Asians, which has negative implications for treatment. It is unclear why this is. MODY should be considered in autoantibody-negative Asian diabetes patients lacking evidence of insulin resistance.

    View details for DOI 10.1111/j.1464-5491.2006.01958.x

    View details for Web of Science ID 000241367000017

    View details for PubMedID 17054605

  • Defining the genetic aetiology monogenic diabetes can improve treatment EXPERT OPINION ON PHARMACOTHERAPY Gloyn, A. L., Ellard, S. 2006; 7 (13): 1759–67

    Abstract

    A molecular genetic diagnosis is now possible for > 80% of patients with monogenic diabetes. This not only provides accurate information regarding inheritance and prognosis, but can inform treatment decisions and improve clinical outcome. Mild fasting hyperglycaemia caused by heterozygous GCK mutations rarely requires pharmacological intervention, whereas patients with mutations in the genes encoding the transcription factors HNF-1alpha and HNF-4alpha respond well to low doses of sulphonylureas. The recent discovery that mutations in the KCNJ11 gene (encoding the Kir6.2 subunit of the K(ATP) channel) are the most common cause of permanent neonatal diabetes, has enabled children to stop insulin injections and achieve improved glycaemic control with high doses of sulphonylurea tablets. Molecular genetic testing is an essential prerequisite for the pharmacogenetic treatment of monogenic diabetes.

    View details for DOI 10.1517/14656566.7.13.1759

    View details for Web of Science ID 000240151300007

    View details for PubMedID 16925503

  • Assessment of the role of common genetic variation in the transient neonatal diabetes mellitus (TNDM) region in type 2 diabetes - A comparative genomic and tagging single nucleotide polymorphism approach DIABETES Gloyn, A. L., Mackay, D. G., Weedon, M. N., McCarthy, M. I., Walker, M., Hitman, G., Knight, B. A., Owen, K. R., Hattersley, A. T., Frayling, T. M. 2006; 55 (8): 2272–76

    Abstract

    Recent evidence supports the strong overlap between genes implicated in monogenic diabetes and susceptibility to type 2 diabetes. Transient neonatal diabetes mellitus (TNDM) is a rare disorder associated with overexpression of genes at a paternally expressed imprinted locus on chromosome 6q24. There are two overlapping genes in this region: the transcription factor zinc finger protein associated with cell cycle control and apoptosis (ZAC also known as PLAGL1) and HYMA1, which encodes an untranslated mRNA. Several type 2 diabetes linkage studies have reported linkage to chromosome 6q22-25. We hypothesized that common genetic variation at this TNDM region influences type 2 diabetes susceptibility. In addition to the coding regions, we used comparative genomic analysis to identify conserved noncoding regions, which were resequenced for single nucleotide polymorphism (SNP) discovery in 47 individuals. Twenty-six SNPs were identified. Fifteen tag SNPs (tSNPs) were successfully genotyped in a large case-control (n = 3,594) and family-based (n = 1,654) study. We did not find any evidence of association or overtransmission of any tSNP to affected offspring or of a parent-of-origin effect. Using a study sufficiently powered to detect odds ratios of <1.2, we conclude that common variation in the TNDM region does not play an important role in the genetic susceptibility to type 2 diabetes.

    View details for DOI 10.2337/db06-0216

    View details for Web of Science ID 000239468300013

    View details for PubMedID 16873690

  • KCNJ11 activating mutations are associated with developmental delay, epilepsy and neonatal diabetes syndrome and other neurological features EUROPEAN JOURNAL OF HUMAN GENETICS Gloyn, A. L., Diatloff-Zito, C., Edghill, E. L., Bellanne-Chantelot, C., Nivot, S., Coutant, R., Ellard, S., Hattersley, A. T., Robert, J. 2006; 14 (7): 824–30

    Abstract

    Heterozygous activating mutations in the gene encoding for the ATP-sensitive potassium channel subunit Kir6.2 (KCNJ11) have recently been shown to be a common cause of permanent neonatal diabetes. Kir6.2 is expressed in muscle, neuron and brain as well as the pancreatic beta-cell, so patients with KCNJ11 mutations could have a neurological phenotype in addition to their diabetes. It is proposed that some patients with KCNJ11 mutations have neurological features that are part of a discrete neurological syndrome termed developmental Delay, Epilepsy and Neonatal Diabetes (DEND), but there are also neurological consequences of chronic or acute diabetes. We identified KCNJ11 mutations in four of 10 probands with permanent neonatal diabetes and one affected parent; this included the novel C166F mutation and the previously described V59M and R201H. Four of the five patients with mutations had neurological features: the patient with the C166F mutation had marked developmental delay, severe generalised epilepsy, hypotonia and muscle weakness; mild developmental delay was present in the patient with the V59M mutation; one patient with the R201H mutation had acute and chronic neurological consequences of cerebral oedema and another had diabetic neuropathy from chronic hyperglycaemia. In conclusion, the clinical features in these patients support the existence of a discrete neurological syndrome with KCNJ11 mutations. The severe DEND syndrome was seen with the novel C166F mutation and mild developmental delay with the V59M mutation. These features differ markedly from the neurological consequences of acute or chronic diabetes.

    View details for DOI 10.1038/sj.ejhg.5201629

    View details for Web of Science ID 000238890100007

    View details for PubMedID 16670688

  • Mutations in KCNJ11, which encodes Kir6.2, are a common cause of diabetes diagnosed in the first 6 months of life, with the phenotype determined by genotype DIABETOLOGIA Flanagan, S. E., Edghill, E. L., Gloyn, A. L., Ellard, S., Hattersley, A. T. 2006; 49 (6): 1190–97

    Abstract

    Heterozygous activating mutations in KCNJ11, which encodes the Kir6.2 subunit of the pancreatic ATP-sensitive potassium (K(ATP)) channel, cause both permanent and transient neonatal diabetes. A minority of patients also have neurological features. The identification of a KCNJ11 mutation has important therapeutic implications, as many patients can replace insulin injections with sulfonylurea tablets. We aimed to determine the age of presentation of patients with KCNJ11 mutations and to examine if there was a relationship between genotype and phenotype.KCNJ11 was sequenced in 239 unrelated patients from 21 countries, who were diagnosed with permanent diabetes before 2 years of age.Thirty-one of the 120 patients (26%) diagnosed in the first 26 weeks of life had a KCNJ11 mutation; no mutations were found in the 119 cases (0%) diagnosed after this age. Fourteen different heterozygous mutations were identified, with the majority resulting from de novo mutations. These include seven novel mutations: H46Y, R50Q, G53D C166Y, K170T, L164P and Y330S. All 11 probands with the most common mutation, R201H, had isolated diabetes. In contrast, developmental delay in addition to diabetes was seen in four of five probands with the V59M mutation and two of four with the R201C mutation. Five patients with developmental delay, epilepsy and neonatal diabetes (DEND) syndrome had unique mutations not associated with other phenotypes.KCNJ11 mutations are a common cause of permanent diabetes diagnosed in the first 6 months and all patients diagnosed in this age group should be tested. There is a strong genotype-phenotype relationship with the mutation being an important determinant of associated neurological features.

    View details for DOI 10.1007/s00125-006-0246-z

    View details for Web of Science ID 000237339700012

    View details for PubMedID 16609879

  • Mutations in the genes encoding the pancreatic beta-cell K-ATP channel subunits Kir6.2 (KCNJ11) and SUR1 (ABCC8) in diabetes mellitus and hyperinsullinlism HUMAN MUTATION Gloyn, A. L., Siddiqui, J., Ellard, S. 2006; 27 (3): 220–31

    Abstract

    The beta-cell ATP-sensitive potassium channel is a key component of stimulus-secretion coupling in the pancreatic beta-cell. The channel couples metabolism to membrane electrical events, bringing about insulin secretion. Given the critical role of this channel in glucose homeostasis, it is not surprising that mutations in the genes encoding for the two essential subunits of the channel can result in both hypo- and hyperglycemia. The channel consists of four subunits of the inwardly rectifying potassium channel Kir6.2 and four subunits of the sulfonylurea receptor 1. It has been known for some time that loss of function mutations in KCNJ11, which encodes for Kir6.2, and ABCC8, which encodes for SUR1, can cause oversecretion of insulin and result in hyperinsulinemia (HI) of infancy; however, heterozygous activating mutations in KCNJ11 that result in the opposite phenotype of diabetes have recently been described. This review focuses on reported mutations in both genes, the spectrum of phenotypes, and the implications for treatment when patients are diagnosed with mutations in these genes.

    View details for DOI 10.1002/humu.20292

    View details for Web of Science ID 000235737800002

    View details for PubMedID 16416420

  • A gating mutation at the internal mouth of the Kir6.2 pore is associated with DEND syndrome EMBO REPORTS Proks, P., Girard, C., Haider, S., Gloyn, A. L., Hattersley, A. T., Sansom, M. S., Ashcroft, F. M. 2005; 6 (5): 470–75

    Abstract

    Inwardly rectifying potassium (Kir) channels control cell membrane K+ fluxes and electrical signalling in diverse cell types. Heterozygous mutations in the human Kir6.2 gene (KCNJ11), the pore-forming subunit of the ATP-sensitive (K(ATP)) channel, cause permanent neonatal diabetes mellitus. However, the I296L mutation also results in developmental delay, muscle weakness and epilepsy. We investigated the functional effects of the I296L mutation by expressing wild-type or mutant Kir6.2/SUR1 channels in Xenopus oocytes. The mutation caused a marked increase in resting whole-cell K(ATP) currents by reducing channel inhibition by ATP, in both homomeric and simulated heterozygous states. Kinetic analysis showed that the mutation impaired ATP sensitivity indirectly, by stabilizing the open state of the channel and possibly also by means of an allosteric effect on ATP binding and/or transduction. The results implicate a new region in Kir-channel gating and suggest that disease severity is correlated with the extent of reduction in ATP sensitivity.

    View details for DOI 10.1038/sj.embor.7400393

    View details for Web of Science ID 000228806300019

    View details for PubMedID 15864298

    View details for PubMedCentralID PMC1299303

  • Relapsing diabetes can result from moderately activating mutations in KCNJ11. Human molecular genetics Gloyn, A. L., Reimann, F. n., Girard, C. n., Edghill, E. L., Proks, P. n., Pearson, E. R., Temple, I. K., Mackay, D. J., Shield, J. P., Freedenberg, D. n., Noyes, K. n., Ellard, S. n., Ashcroft, F. M., Gribble, F. M., Hattersley, A. T. 2005; 14 (7): 925–34

    Abstract

    Neonatal diabetes can either remit and hence be transient or else may be permanent. These two phenotypes were considered to be genetically distinct. Abnormalities of 6q24 are the commonest cause of transient neonatal diabetes (TNDM). Mutations in KCNJ11, which encodes Kir6.2, the pore-forming subunit of the ATP-sensitive potassium channel (K(ATP)), are the commonest cause of permanent neonatal diabetes (PNDM). In addition to diabetes, some KCNJ11 mutations also result in marked developmental delay and epilepsy. These mutations are more severe on functional characterization. We investigated whether mutations in KCNJ11 could also give rise to TNDM. We identified the three novel heterozygous mutations (G53S, G53R, I182V) in three of 11 probands with clinically defined TNDM, who did not have chromosome 6q24 abnormalities. The mutations co-segregated with diabetes within families and were not found in 100 controls. All probands had insulin-treated diabetes diagnosed in the first 4 months and went into remission by 7-14 months. Functional characterization of the TNDM associated mutations was performed by expressing the mutated Kir6.2 with SUR1 in Xenopus laevis oocytes. All three heterozygous mutations resulted in a reduction in the sensitivity to ATP when compared with wild-type (IC(50) approximately 30 versus approximately 7 microM, P-value for is all <0.01); however, this was less profoundly reduced than with the PNDM associated mutations. In conclusion, mutations in KCNJ11 are the first genetic cause for remitting as well as permanent diabetes. This suggests that a fixed ion channel abnormality can result in a fluctuating glycaemic phenotype. The multiple phenotypes associated with activating KCNJ11 mutations may reflect their severity in vitro.

    View details for DOI 10.1093/hmg/ddi086

    View details for PubMedID 15718250

  • KCNJ11 activating mutations in Italian patients with permanent neonatal diabetes HUMAN MUTATION Massa, O., Iafusco, D., D'Amato, E., Gloyn, A. L., Hattersley, A. T., Pasquino, B., Tonini, G., Dammacco, F., Zanette, G., Meschi, F., Porzio, O., Bottazzo, G., Crino, A., Lorini, R., Cerutti, F., Vanelli, M., Barbetti, F., Early Onset Diabet Study Grp Itali 2005; 25 (1): 22–27

    Abstract

    Permanent neonatal diabetes mellitus (PNDM) is a rare condition characterized by severe hyperglycemia constantly requiring insulin treatment from its onset. Complete deficiency of glucokinase (GCK) can cause PNDM; however, the genetic etiology is unknown in most PNDM patients. Recently, heterozygous activating mutations of KCNJ11, encoding Kir6.2, the pore forming subunit of the ATP-dependent potassium (K(ATP)) channel of the pancreatic beta-cell, were found in patients with PNDM. Closure of the K(ATP) channel exerts a pivotal role in insulin secretion by modifying the resting membrane potential that leads to insulin exocytosis. We screened the KCNJ11 gene in 12 Italian patients with PNDM (onset within 3 months from birth) and in six patients with non-autoimmune, insulin-requiring diabetes diagnosed during the first year of life. Five different heterozygous mutations were identified: c.149G>C (p.R50P), c.175G>A (p.V59M), c.509A>G (p.K170R), c.510G>C (p.K170N), and c.601C>T (p.R201C) in eight patients with diabetes diagnosed between day 3 and 182. Mutations at Arg50 and Lys170 residues are novel. Four patients also presented with motor and/or developmental delay as previously reported. We conclude that KCNJ11 mutations are a common cause of PNDM either in isolation or associated with developmental delay. Permanent diabetes of non autoimmune origin can present up to 6 months from birth in individuals with KCNJ11 and EIF2AK3 mutations. Therefore, we suggest that the acronym PNDM be replaced with the more comprehensive permanent diabetes mellitus of infancy (PDMI), linking it to the gene product (e.g., GCK-PDMI, KCNJ11-PDMI) to avoid confusion between patients with early-onset, autoimmune type 1 diabetes.

    View details for DOI 10.1002/humu.20124

    View details for Web of Science ID 000226018800004

    View details for PubMedID 15580558

  • Permanent neonatal diabetes in an Asian infant JOURNAL OF PEDIATRICS Porter, Shaw, N. J., Barrett, T. G., Hattersley, A. T., Ellard, S., Gloyn, A. L. 2005; 146 (1): 131–33

    Abstract

    We describe a novel homozygous missense glucokinase mutation (R397L) resulting in insulin-treated neonatal diabetes in an infant from a consanguineous Asian family. Both parents were heterozygous for R397L and had mild hyperglycemia. Glucokinase mutations should be considered in infants of all ethnic groups with neonatal diabetes and consanguinity.

    View details for DOI 10.1016/j.jpeds.2004.09.008

    View details for Web of Science ID 000226337700031

    View details for PubMedID 15644838

  • Insights into the structure and regulation of glucokinase from a novel mutation (V62M), which causes maturity-onset diabetes of the young. The Journal of biological chemistry Gloyn, A. L., Odili, S. n., Zelent, D. n., Buettger, C. n., Castleden, H. A., Steele, A. M., Stride, A. n., Shiota, C. n., Magnuson, M. A., Lorini, R. n., d'Annunzio, G. n., Stanley, C. A., Kwagh, J. n., van Schaftingen, E. n., Veiga-da-Cunha, M. n., Barbetti, F. n., Dunten, P. n., Han, Y. n., Grimsby, J. n., Taub, R. n., Ellard, S. n., Hattersley, A. T., Matschinsky, F. M. 2005; 280 (14): 14105–13

    Abstract

    Glucokinase (GCK) serves as the pancreatic glucose sensor. Heterozygous inactivating GCK mutations cause hyperglycemia, whereas activating mutations cause hypoglycemia. We studied the GCK V62M mutation identified in two families and co-segregating with hyperglycemia to understand how this mutation resulted in reduced function. Structural modeling locates the mutation close to five naturally occurring activating mutations in the allosteric activator site of the enzyme. Recombinant glutathionyl S-transferase-V62M GCK is paradoxically activated rather than inactivated due to a decreased S0.5 for glucose compared with wild type (4.88 versus 7.55 mM). The recently described pharmacological activator (RO0281675) interacts with GCK at this site. V62M GCK does not respond to RO0281675, nor does it respond to the hepatic glucokinase regulatory protein (GKRP). The enzyme is also thermally unstable, but this lability is apparently less pronounced than in the proven instability mutant E300K. Functional and structural analysis of seven amino acid substitutions at residue Val62 has identified a non-linear relationship between activation by the pharmacological activator and the van der Waals interactions energies. Smaller energies allow a hydrophobic interaction between the activator and glucokinase, whereas larger energies prohibit the ligand from fitting into the binding pocket. We conclude that V62M may cause hyperglycemia by a complex defect of GCK regulation involving instability in combination with loss of control by a putative endogenous activator and/or GKRP. This study illustrates that mutations that cause hyperglycemia are not necessarily kinetically inactivating but may exert their effects by other complex mechanisms. Elucidating such mechanisms leads to a deeper understanding of the GCK glucose sensor and the biochemistry of beta-cells and hepatocytes.

    View details for DOI 10.1074/jbc.M413146200

    View details for PubMedID 15677479

  • Molecular basis of Kir6.2 mutations associated with neonatal diabetes or neonatal diabetes plus neurological features PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Proks, P., Antcliff, J. F., Lippiat, J., Gloyn, A. L., Hattersley, A. T., Ashcroft, F. M. 2004; 101 (50): 17539–44

    Abstract

    Inwardly rectifying potassium channels (Kir channels) control cell membrane K(+) fluxes and electrical signaling in diverse cell types. Heterozygous mutations in the human Kir6.2 gene (KCNJ11), the pore-forming subunit of the ATP-sensitive (K(ATP)) channel, cause permanent neonatal diabetes mellitus (PNDM). For some mutations, PNDM is accompanied by marked developmental delay, muscle weakness, and epilepsy (severe disease). To determine the molecular basis of these different phenotypes, we expressed wild-type or mutant (R201C, Q52R, or V59G) Kir6.2/sulfonylurea receptor 1 channels in Xenopus oocytes. All mutations increased resting whole-cell K(ATP) currents by reducing channel inhibition by ATP, but, in the simulated heterozygous state, mutations causing PNDM alone (R201C) produced smaller K(ATP) currents and less change in ATP sensitivity than mutations associated with severe disease (Q52R and V59G). This finding suggests that increased K(ATP) currents hyperpolarize pancreatic beta cells and impair insulin secretion, whereas larger K(ATP) currents are required to influence extrapancreatic cell function. We found that mutations causing PNDM alone impair ATP sensitivity directly (at the binding site), whereas those associated with severe disease act indirectly by biasing the channel conformation toward the open state. The effect of the mutation on ATP sensitivity in the heterozygous state reflects the different contributions of a single subunit in the Kir6.2 tetramer to ATP inhibition and to the energy of the open state. Our results also show that mutations in the slide helix of Kir6.2 (V59G) influence the channel kinetics, providing evidence that this domain is involved in Kir channel gating, and suggest that the efficacy of sulfonylurea therapy in PNDM may vary with genotype.

    View details for DOI 10.1073/pnas.0404756101

    View details for Web of Science ID 000225803400042

    View details for PubMedID 15583126

    View details for PubMedCentralID PMC536014

  • Mutations in PTF1A cause pancreatic and cerebellar agenesis NATURE GENETICS Sellick, G. S., Barker, K. T., Stolte-Dijkstra, Fleischmann, C., Coleman, R. J., Garrett, C., Gloyn, A. L., Edghill, E. L., Hattersley, A. T., Wellauer, P. K., Goodwin, G., Houlston, R. S. 2004; 36 (12): 1301–5

    Abstract

    Individuals with permanent neonatal diabetes mellitus usually present within the first three months of life and require insulin treatment. We recently identified a locus on chromosome 10p13-p12.1 involved in permanent neonatal diabetes mellitus associated with pancreatic and cerebellar agenesis in a genome-wide linkage search of a consanguineous Pakistani family. Here we report the further linkage analysis of this family and a second family of Northern European descent segregating an identical phenotype. Positional cloning identified the mutations 705insG and C886T in the gene PTF1A, encoding pancreas transcription factor 1alpha, as disease-causing sequence changes. Both mutations cause truncation of the expressed PTF1A protein C-terminal to the basic-helix-loop-helix domain. Reporter-gene studies using a minimal PTF1A deletion mutant indicate that the deleted region defines a new domain that is crucial for the function of this protein. PTF1A is known to have a role in mammalian pancreatic development, and the clinical phenotype of the affected individuals implicated the protein as a key regulator of cerebellar neurogenesis. The essential role of PTF1A in normal cerebellar development was confirmed by detailed neuropathological analysis of Ptf1a(-/-) mice.

    View details for DOI 10.1038/ng1475

    View details for Web of Science ID 000225354100018

    View details for PubMedID 15543146

  • Permanent neonatal diabetes due to mutations in KCNJ11 encoding Kir6.2 - Patient characteristics and initial response to sulfonylurea therapy DIABETES Sagen, J. V., Raeder, H., Hathout, E., Shehadeh, N., Gudmundsson, K., Baevre, H., Abuelo, D., Phornphutkul, C., Molnes, J., Bell, G. I., Gloyn, A. L., Hattersley, A. T., Molven, A., Sovik, O., Njolstad, P. R. 2004; 53 (10): 2713–18

    Abstract

    Permanent neonatal diabetes (PND) can be caused by mutations in the transcription factors insulin promoter factor (IPF)-1, eukaryotic translation initiation factor-2alpha kinase 3 (EIF2AK3), and forkhead box-P3 and in key components of insulin secretion: glucokinase (GCK) and the ATP-sensitive K(+) channel subunit Kir6.2. We sequenced the gene encoding Kir6.2 (KCNJ11) in 11 probands with GCK-negative PND. Heterozygous mutations were identified in seven probands, causing three novel (F35V, Y330C, and F333I) and two known (V59M and R201H) Kir6.2 amino acid substitutions. Only two probands had a family history of diabetes. Subjects with the V59M mutation had neurological features including motor delay. Three mutation carriers tested had an insulin secretory response to tolbutamide, but not to glucose or glucagon. Glibenclamide was introduced in increasing doses to investigate whether sulfonylurea could replace insulin. At a glibenclamide dose of 0.3-0.4 mg. kg(-1). day(-1), insulin was discontinued. Blood glucose did not deteriorate, and HbA(1c) was stable or fell during 2-6 months of follow-up. An oral glucose tolerance test performed in one subject revealed that glucose-stimulated insulin release was restored. Mutations in Kir6.2 were the most frequent cause of PND in our cohort. Apparently insulin-dependent patients with mutations in Kir6.2 may be managed on an oral sulfonylurea with sustained metabolic control rather than insulin injections, illustrating the principle of pharmacogenetics applied in diabetes treatment.

    View details for DOI 10.2337/diabetes.53.10.2713

    View details for Web of Science ID 000224116100028

    View details for PubMedID 15448106

  • Kir6.2 mutations are a common cause of permanent neonatal diabetes in a large cohort of French patients DIABETES Vaxillaire, M., Populaire, C., Busiah, K., Cave, H., Gloyn, A. L., Hattersley, A. T., Czernichow, P., Froguel, P., Polak, M. 2004; 53 (10): 2719–22

    Abstract

    Permanent neonatal diabetes (PND), requiring insulin within the first months of life, is unexplained at the molecular level in most cases. It has very recently been shown that heterozygous activating mutations in the KCNJ11 gene, encoding the Kir6.2 subunit of the pancreatic ATP-sensitive K(+) channel involved in the regulation of insulin secretion, cause PND. In the present study, we screened the KCNJ11 gene for mutations in French patients with PND. Patients were recruited through the French network for the study of neonatal diabetes. Seventeen at-term babies with a median age at diagnosis of diabetes of 64 days (range 1-260) were included. We identified in nine patients seven heterozygous nonsynonymous mutations: three of them (V59M, R201C, and R201H) were already described, and the four novel mutations resulted in an amino acid change of Kir6.2 at positions F35L, G53N, E322K, and Y330C. More patients with a Kir6.2 mutation (six of nine) were reported to have a smaller birth weight than those without mutation (two of eight). Although Kir6.2 mutation carriers do not represent a phenotypically specific form of PND, an impaired function of Kir6.2 is associated with in utero insulin secretory insufficiency and growth retardation. In conclusion, we confirmed that Kir6.2 mutations are a common cause (53%) of PND in Caucasians.

    View details for DOI 10.2337/diabetes.53.10.2719

    View details for Web of Science ID 000224116100029

    View details for PubMedID 15448107

  • Permanent neonatal diabetes due to paternal germline mosaicism for an activating mutation of the KCNJ11 gene encoding the Kir6.2 subunit of the beta-cell potassium adenosine triphosphate channel JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM Gloyn, A. L., Cummings, E. A., Edghill, E. L., Harries, L. W., Scott, R., Costa, T., Temple, I. K., Hattersley, A. T., Ellard, S. 2004; 89 (8): 3932–35

    Abstract

    Activating mutations in the KCNJ11 gene encoding for the Kir6.2 subunit of the beta-cell ATP-sensitive potassium channel have recently been shown to be a common cause of permanent neonatal diabetes. In 80% of probands, these are isolated cases resulting from de novo mutations. We describe a family in which two affected paternal half-siblings were found to be heterozygous for the previously reported R201C mutation. Direct sequencing of leukocyte DNA showed that their clinically unaffected mothers and father were genotypically normal. Quantitative real-time PCR analysis of the father's leukocyte DNA detected no trace of mutant DNA. These results are consistent with the father being a mosaic for the mutation, which is restricted to his germline. This is the first report of germline mosaicism in any form of monogenic diabetes. The high percentage of permanent neonatal diabetes cases due to de novo KCNJ11 mutations suggests that germline mosaicism may be common. The possibility of germline mosaicism should be considered when counseling recurrence risks for the parents of a child with an apparently de novo KCNJ11 activating mutation.

    View details for DOI 10.1210/jc.2004-0568

    View details for Web of Science ID 000223072400051

    View details for PubMedID 15292329

  • Activating mutations in the KCNJ11 gene encoding the ATP-sensitive K+ channel subunit Kir6.2 are rare in clinically defined type 1 diabetes diagnosed before 2 years. Diabetes Edghill, E. L., Gloyn, A. L., Gillespie, K. M., Lambert, A. P., Raymond, N. T., Swift, P. G., Ellard, S. n., Gale, E. A., Hattersley, A. T. 2004; 53 (11): 2998–3001

    Abstract

    We have recently shown that permanent neonatal diabetes can be caused by activating mutations in KCNJ11 that encode the Kir6.2 subunit of the beta-cell ATP-sensitive K(+) channel. Some of these patients were diagnosed after 3 months of age and presented with ketoacidosis and marked hyperglycemia, which could have been diagnosed as type 1 diabetes. We hypothesized that KCNJ11 mutations could present clinically as type 1 diabetes. We screened the KCNJ11 gene for mutations in 77 U.K. type 1 diabetic subjects diagnosed under the age of 2 years. One patient was found to be heterozygous for the missense mutation R201C. She had low birth weight, was diagnosed at 5 weeks, and did not have a high risk predisposing HLA genotype. A novel variant, R176C, was identified in one diabetic subject but did not cosegregate with diabetes within the family. In conclusion, we have shown that heterozygous activating mutations in the KCNJ11 gene are a rare cause of clinically defined type 1 diabetes diagnosed before 2 years. Although activating KCNJ11 mutations are rare in patients diagnosed with type 1 diabetes, the identification of a KCNJ11 mutation may have important treatment implications.

    View details for DOI 10.2337/diabetes.53.11.2998

    View details for PubMedID 15504982

  • Activating mutations in the gene encoding the ATP-sensitive potassium-channel subunit Kir6.2 and permanent neonatal diabetes. The New England journal of medicine Gloyn, A. L., Pearson, E. R., Antcliff, J. F., Proks, P. n., Bruining, G. J., Slingerland, A. S., Howard, N. n., Srinivasan, S. n., Silva, J. M., Molnes, J. n., Edghill, E. L., Frayling, T. M., Temple, I. K., Mackay, D. n., Shield, J. P., Sumnik, Z. n., van Rhijn, A. n., Wales, J. K., Clark, P. n., Gorman, S. n., Aisenberg, J. n., Ellard, S. n., Njølstad, P. R., Ashcroft, F. M., Hattersley, A. T. 2004; 350 (18): 1838–49

    Abstract

    Patients with permanent neonatal diabetes usually present within the first three months of life and require insulin treatment. In most, the cause is unknown. Because ATP-sensitive potassium (K(ATP)) channels mediate glucose-stimulated insulin secretion from the pancreatic beta cells, we hypothesized that activating mutations in the gene encoding the Kir6.2 subunit of this channel (KCNJ11) cause neonatal diabetes.We sequenced the KCNJ11 gene in 29 patients with permanent neonatal diabetes. The insulin secretory response to intravenous glucagon, glucose, and the sulfonylurea tolbutamide was assessed in patients who had mutations in the gene.Six novel, heterozygous missense mutations were identified in 10 of the 29 patients. In two patients the diabetes was familial, and in eight it arose from a spontaneous mutation. Their neonatal diabetes was characterized by ketoacidosis or marked hyperglycemia and was treated with insulin. Patients did not secrete insulin in response to glucose or glucagon but did secrete insulin in response to tolbutamide. Four of the patients also had severe developmental delay and muscle weakness; three of them also had epilepsy and mild dysmorphic features. When the most common mutation in Kir6.2 was coexpressed with sulfonylurea receptor 1 in Xenopus laevis oocytes, the ability of ATP to block mutant K(ATP) channels was greatly reduced.Heterozygous activating mutations in the gene encoding Kir6.2 cause permanent neonatal diabetes and may also be associated with developmental delay, muscle weakness, and epilepsy. Identification of the genetic cause of permanent neonatal diabetes may facilitate the treatment of this disease with sulfonylureas.

    View details for DOI 10.1056/NEJMoa032922

    View details for PubMedID 15115830

  • Genetics for Endocrinologists: The Molecular Genetic Basis of Endocrine Disorders. Clinical endocrinology McCarthy, M. I., Gloyn, A. L. 2003; 59 (6): 826

    View details for DOI 10.1046/j.1365-2265.2003.01927.x

    View details for PubMedID 14974932

  • Identification of 21 novel glucokinase (GCK) mutations in UK and European Caucasians with maturity-onset diabetes of the young (MODY). Human mutation Thomson, K. L., Gloyn, A. L., Colclough, K., Batten, M., Allen, L. I., Beards, F., Hattersley, A. T., Ellard, S. 2003; 22 (5): 417

    Abstract

    Maturity-onset diabetes of the young (MODY) resulting from mutations in the glucokinase (GCK) gene accounts for approximately 20% of MODY in the UK. We have performed fluorescent single stranded conformation polymorphism (F-SSCP) analysis or direct sequencing of the GCK gene in 212 patients referred as part of a research cohort or for diagnostic molecular genetic testing. Mutation screening has identified 43 different mutations in 61 individuals, of which 21 are novel. This report details the mutations identified and their associated clinical features.

    View details for DOI 10.1002/humu.9186

    View details for PubMedID 14517956

  • Quantitative traits associated with the Type 2 diabetes susceptibility allele in Kir6.2 DIABETOLOGIA Weedon, M. N., Gloyn, A. L., Frayling, T. M., Hattersley, A. T., Smith, G. D., Ben-Shlomo, Y. 2003; 46 (7): 1021-1023

    View details for DOI 10.1007/s00125-003-1135-3

    View details for Web of Science ID 000184644300021

    View details for PubMedID 12819904

  • The search for type 2 diabetes genes AGEING RESEARCH REVIEWS Gloyn, A. L. 2003; 2 (2): 111–27

    Abstract

    Type 2 diabetes (T2DM) is a serious disease with severe complications. Around one in 10 people alive today suffer from type 2 diabetes or are destined to develop it before they die. Inheritance plays an important role in the cause of type 2 diabetes. A considerable amount of research is devoted to defining the genes involved in the aetiology of this widespread disease. This information is crucial if we are to improve our methods of preventing and treating diabetes. Over the last 25 years there have been considerable advances in our understanding of the genetics of diabetes. Important discoveries have been made in dissecting the genes involved in rare monogenic forms of type 2 diabetes which has become a paradigm for genetic studies of type 2 diabetes. This review focuses on the main approaches currently adopted and our current understanding of the genes involved in susceptibility to type 2 diabetes.

    View details for DOI 10.1016/S1568-1637(02)00061-2

    View details for Web of Science ID 000183599600001

    View details for PubMedID 12605956

  • Glucokinase (GCK) mutations in hyper- and hypoglycemia: maturity-onset diabetes of the young, permanent neonatal diabetes, and hyperinsulinemia of infancy. Human mutation Gloyn, A. L. 2003; 22 (5): 353–62

    Abstract

    Glucokinase is a key regulatory enzyme in the pancreatic beta-cell. It plays a crucial role in the regulation of insulin secretion and has been termed the pancreatic beta-cell sensor. Given its central role in the regulation of insulin release, it is understandable that mutations in the gene encoding glucokinase (GCK) can cause both hyperglycemia and hypoglycemia. Heterozygous inactivating mutations in GCK cause maturity-onset diabetes of the young (MODY), characterized by mild hyperglycemia, which is present at birth, but is often only detected later in life during screening for other purposes. Homozygous inactivating GCK mutations result in a more severe phenotype, presenting at birth as permanent neonatal diabetes mellitus (PNDM). Several heterozygous activating GCK mutations that cause hypoglycemia have also been reported. A total of 195 mutations in the GCK gene have been described, in a total of 285 families. There are no common mutations and the mutations are distributed throughout the gene. Mutations that cause hypoglycemia are located in various exons in a discrete region of the protein termed the heterotropic allosteric activator site. The identification of a GCK mutation in hyper- and hypoglycemia has implications for the clinical course and clinical management of the disorder.

    View details for DOI 10.1002/humu.10277

    View details for PubMedID 14517946

  • Insights into the biochemical and genetic basis of glucokinase activation from naturally occurring hypoglycemia mutations. Diabetes Gloyn, A. L., Noordam, K. n., Willemsen, M. A., Ellard, S. n., Lam, W. W., Campbell, I. W., Midgley, P. n., Shiota, C. n., Buettger, C. n., Magnuson, M. A., Matschinsky, F. M., Hattersley, A. T. 2003; 52 (9): 2433–40

    Abstract

    Glucokinase (GCK) is a key regulatory enzyme in the pancreatic beta-cell and catalyzes the rate-limiting step for beta-cell glucose metabolism. We report two novel GCK mutations (T65I and W99R) that have arisen de novo in two families with familial hypoglycemia. Insulin levels, although inappropriately high for the degree of hypoglycemia, remain regulated by fluctuations in glycemia, and pancreatic histology was normal. These mutations are within the recently identified heterotropic allosteric activator site in the theoretical model of human beta-cell glucokinase. Functional analysis of the purified recombinant glutathionyl S-transferase fusion proteins of T65I and W99R GCK revealed that the kinetic changes result in a relative increased activity index (a measure of the enzyme's phosphorylating potential) of 9.81 and 6.36, respectively, compared with wild-type. The predicted thresholds for glucose-stimulated insulin release using mathematical modeling were 3.1 (T65I) and 2.8 (W99R) mmol/l, which were in line with the patients' fasting glucose. In conclusion, we have identified two novel spontaneous GCK-activating mutations whose clinical phenotype clearly differs from mutations in ATP-sensitive K(+) channel genes. In vitro studies confirm the validity of structural and functional models of GCK and the putative allosteric activator site, which is a potential drug target for the treatment of type 2 diabetes.

    View details for DOI 10.2337/diabetes.52.9.2433

    View details for PubMedID 12941786

  • Large-scale association studies of variants in genes encoding the pancreatic beta-cell KATP channel subunits Kir6.2 (KCNJ11) and SUR1 (ABCC8) confirm that the KCNJ11 E23K variant is associated with type 2 diabetes. Diabetes Gloyn, A. L., Weedon, M. N., Owen, K. R., Turner, M. J., Knight, B. A., Hitman, G. n., Walker, M. n., Levy, J. C., Sampson, M. n., Halford, S. n., McCarthy, M. I., Hattersley, A. T., Frayling, T. M. 2003; 52 (2): 568–72

    Abstract

    The genes ABCC8 and KCNJ11, which encode the subunits sulfonylurea receptor 1 (SUR1) and inwardly rectifying potassium channel (Kir6.2) of the beta-cell ATP-sensitive potassium (K(ATP)) channel, control insulin secretion. Common polymorphisms in these genes (ABCC8 exon 16-3t/c, exon 18 T/C, KCNJ11 E23K) have been variably associated with type 2 diabetes, but no large ( approximately 2,000 subjects) case-control studies have been performed. We evaluated the role of these three variants by studying 2,486 U.K. subjects: 854 with type 2 diabetes, 1,182 population control subjects, and 150 parent-offspring type 2 diabetic trios. The E23K allele was associated with diabetes in the case-control study (odds ratio [OR] 1.18 [95% CI 1.04-1.34], P = 0.01) but did not show familial association with diabetes. Neither the exon 16 nor the exon 18 ABCC8 variants were associated with diabetes (1.04 [0.91-1.18], P = 0.57; 0.93 [0.71-1.23], P = 0.63, respectively). Meta-analysis of all case-control data showed that the E23K allele was associated with type 2 diabetes (K allele OR 1.23 [1.12-1.36], P = 0.000015; KK genotype 1.65 [1.34-2.02], P = 0.000002); but the ABCC8 variants were not associated. Our results confirm that E23K increases risk of type 2 diabetes and show that large-scale association studies are important for the identification of diabetes susceptibility alleles.

    View details for DOI 10.2337/diabetes.52.2.568

    View details for PubMedID 12540637

  • A putative functional polymorphism in the IGF-I gene - Association studies with type 2 diabetes, adult height, glucose tolerance, and fetal growth in UK populations DIABETES Frayling, T. M., Hattersley, A. T., McCarthy, A., Holly, J., Mitchell, S. M., Gloyn, A. L., Owen, K., Davies, D., Smith, G. D., Ben-Shlomo, Y. 2002; 51 (7): 2313–16

    Abstract

    IGF-I has a critical role in growth and metabolism. A microsatellite polymorphism 1 kb upstream to the IGF-I gene has recently been associated with several adult phenotypes. In a large Dutch cohort, the absence of the commonest allele (Z) was associated with reduced serum IGF-I levels, reduced height, and an increased risk of type 2 diabetes and myocardial infarction. This result has not been replicated, and the role of this polymorphism in these traits in U.K. subjects is not known. We sought further evidence for the involvement of this variant in type 2 diabetes using a case-control study and IGF-I and diabetes-related traits in a population cohort of 640 U.K. individuals aged 25 years. Absence of the common allele was not associated with type 2 diabetes (odds ratio 0.70, 95% CI 0.47-1.04 for X/X versus Z/Z genotype, chi(2) test for trend across genotypes, P = 0.018). In the population cohort, the common allele (Z) was associated with decreased IGF-I levels (P = 0.01), contrary to the Dutch study, but not with adult height (P = 0.23), glucose tolerance (P = 0.84), oral glucose tolerance test-derived values of beta-cell function (P = 0.90), or insulin resistance (P = 0.66). There was no association with measures of fetal growth, including birth weight (P = 0.17). Our results do not support the previous associations and suggest that the promoter microsatellite is unlikely to be functionally important.

    View details for DOI 10.2337/diabetes.51.7.2313

    View details for Web of Science ID 000176616200042

    View details for PubMedID 12086966

  • The role of the HNF4 alpha enhancer in type 2 diabetes variants of the HNF4 alpha enhancer are not a common cause of susceptibility to type 2 diabetes MOLECULAR GENETICS AND METABOLISM Mitchell, S. M., Gloyn, A. L., Owen, K. R., Hattersley, A. T., Frayling, T. M. 2002; 76 (2): 148–51

    Abstract

    The genetic causes of type 2 diabetes are not well understood. The disease has been linked to chromosome 20q12-q13.1 a region which harbors the transcription factor HNF4alpha. Mutations in the coding region of HNF4alpha cause maturity onset diabetes of the young, an autosomal dominant form of diabetes, but do not account for the linkage to this region. An enhancer element has recently been characterized 6 kb 5' of the HNF4alpha P1 promoter containing binding sites for the transcription factors HNF1, HNF4, HNF3, and C/EBP, which are overlapped by glucocorticoid consensus sites. We hypothesized that variation in the enhancer element disrupts HNF4alpha expression in the liver and increases susceptibility to type 2 diabetes. We screened for variants of the enhancer element in 39 white UK young onset diabetic subjects, giving >95% power to identify variants with minor allele frequencies of >5%. No variants of the enhancer element were found in this population. We conclude that variation in the HNF4alpha enhancer element is not a common cause of susceptibility to type 2 diabetes.

    View details for DOI 10.1016/S1096-7192(02)00027-6

    View details for Web of Science ID 000176747600009

    View details for PubMedID 12083813

  • Maturity-onset diabetes of the young caused by a balanced translocation where the 20q12 break point results in disruption upstream of the coding region of hepatocyte nuclear factor-4alpha (HNF4A) gene. Diabetes Gloyn, A. L., Ellard, S. n., Shepherd, M. n., Howell, R. T., Parry, E. M., Jefferson, A. n., Levy, E. R., Hattersley, A. T. 2002; 51 (7): 2329–33

    Abstract

    Monogenic human disorders have been used as paradigms for complex genetic disease and as tools for establishing important insights into mechanisms of gene regulation and transcriptional control. Maturity-onset diabetes of the young (MODY) is a monogenic dominantly inherited form of diabetes that is characterized by defective insulin secretion from the pancreatic beta-cells. A wide variety of mutation types in five different genes have been identified that result in this condition. There have been no reports of a chromosome deletion or translocation resulting in MODY. We report a pedigree where MODY cosegregates with a balanced translocation [karyotype 46, XX t(3;20) (p21.2;q12)]. The chromosome 20 break point, 20q12, is within the region of one of the known MODY genes, hepatocyte nuclear factor-4alpha (HNF4A). Fluorescence in situ hybridization analysis demonstrated that the break point does not disrupt the coding region of this gene, but it lies at least 6 kb upstream of the conventional promoter (P1). We propose that this mutation disrupts the spatial relationship between the recently described alternate distal pancreatic promoter (P2) and HNF4A. This is the first case of MODY due to a balanced translocation, and it provides evidence to confirm the crucial role of an upstream regulator of HNF4A gene expression in the beta-cell.

    View details for DOI 10.2337/diabetes.51.7.2329

    View details for PubMedID 12086970

  • Human calcium/calmodulin-dependent protein kinase II gamma gene (CAMK2G): cloning, genomic structure and detection of variants in subjects with type II diabetes. Diabetologia Gloyn, A. L., Desai, M. n., Clark, A. n., Levy, J. C., Holman, R. R., Frayling, T. M., Hattersley, A. T., Ashcroft, S. J. 2002; 45 (4): 580–83

    Abstract

    Ca(2+)/calmodulin-dependent protein kinase II, is expressed in the pancreatic beta cells and is activated by glucose and other secretagogues in a manner correlating with insulin secretion. The activation of Ca(2+)/calmodulin-dependent protein kinase II mediates some of the actions of Ca(2+) on the exocytosis of insulin. We therefore investigated the gene encoding the gamma isoform ( CAMK2G) which has been shown to be expressed in human beta cells as a candidate gene for Type II (non-insulin-dependent) diabetes mellitus.Human CAMK2G was cloned from a total human P1 artificial chromosome library using a partial Ca(2+)/calmodulin-dependent protein kinase gamma(E) cDNA probe. Positive PAC clones were localised to chromosome 10q22 by fluorescence in situ hybridisation. To obtain structural information and the sequences of the exon-intron boundaries, the published genomic structures of the rat and mouse genes allowed the putative exon-intron boundaries of human CAMK2G to be amplified by vectorette polymerase chain reaction and sequenced. Sequence variants in each exon were identified using single stranded conformational polymorphism analysis.The human CAMK2G gene comprises 22 exons which range in size between 43 to 230 bp. Screening of the exons and exon-intron boundaries identified two single nucleotide polymorphisms. These did not show association with diabetes in 122 patients and 144 control subjects.We have identified the genomic structure of CAMK2G to enable further study of this potential candidate gene. Variation in this gene is not strongly associated with diabetes in Caucasians in the United Kingdom. We have identified two single nucleotide polymorphisms which, with appropriately large case control studies, can be used to assess the role of CAMK2G in the susceptibility to Type II diabetes.

    View details for DOI 10.1007/s00125-002-0779-8

    View details for PubMedID 12032636

  • Complete glucokinase deficiency is not a common cause of permanent neonatal diabetes. Diabetologia Gloyn, A. L., Ellard, S. n., Shield, J. P., Temple, I. K., Mackay, D. J., Polak, M. n., Barrett, T. n., Hattersley, A. T. 2002; 45 (2): 290

    View details for DOI 10.1007/s00125-001-0746-9

    View details for PubMedID 11942315

  • The genetics of type 2 diabetes BEST PRACTICE & RESEARCH CLINICAL ENDOCRINOLOGY & METABOLISM Gloyn, A. L., McCarthy, M. I. 2001; 15 (3): 293–308

    Abstract

    Type 2 diabetes mellitus is not a single disease but a genetically heterogeneous group of metabolic disorders sharing glucose intolerance. The precise underlying biochemical defects are unknown and almost certainly include impairments of both insulin secretion and action. The rapidly increasing prevalence of T2D world wide makes it a major cause of morbidity and mortality. Understanding the genetic aetiology of T2D will facilitate its diagnosis, treatment and prevention. The results of linkage and association studies to date demonstrate that, as with other common diseases, multiple genes are involved in the susceptibility to T2D, each making a modest contribution to the overall risk. The completion of the draft human genome sequence and a brace of novel tools for genomic analysis promise to accelerate progress towards a more complete molecular description of T2D.

    View details for DOI 10.1053/beem.2001.0147

    View details for Web of Science ID 000171229800004

    View details for PubMedID 11554772

  • The beta-cell Ca2+/calmodulin-dependent protein kinase II (CaM kinase II) beta3 isoform containing a proline-rich tandem repeat in the association domain can be found in the human genome. Diabetologia Gloyn, A. L., Ashcroft, S. J. 2001; 44 (6): 787

    View details for DOI 10.1007/s001250051691

    View details for PubMedID 11440377

  • Association studies of variants in promoter and coding regions of beta-cell ATP-sensitive K-channel genes SUR1 and Kir6.2 with Type 2 diabetes mellitus (UKPDS 53). Diabetic medicine : a journal of the British Diabetic Association Gloyn, A. L., Hashim, Y. n., Ashcroft, S. J., Ashfield, R. n., Wiltshire, S. n., Turner, R. C. 2001; 18 (3): 206–12

    Abstract

    The beta-cell ATP-sensitive potassium channel consists of two subunits, SUR1 and Kir6.2. Population association studies have shown that three variants in SUR1 and one in Kir6.2 are associated with Type 2 diabetes. These polymorphisms do not result in a functional change or affect splicing, suggesting that they could be in linkage disequilibrium with a pathogenic mutation. The present study aimed firstly to screen the promoter regions of SUR1 and Kir6.2 to determine whether mutations in linkage disequilibrium with the silent variants lie in regulatory regions, which might lead to changes in gene expression. Secondly, novel and previously described variants associated with Type 2 diabetes (SUR1 exon 16-3t, exon 18 T, and Kir6.2 E23K) were investigated in the UKPDS cohort.The promoter sequences of both genes were screened by single-stranded conformational polymorphism analysis for variants associated with Type 2 diabetes. The previously reported variants were evaluated in 364 Type 2 diabetic and 328 normoglycaemic control subjects.Two variants were detected in the SUR1 promoter, a three base insertion (caa) at -522 bp and a single base substitution at - 679 bp (c-->g). Neither of the variants were associated with diabetes, nor were they in a sequence consensus region for transcription factors. No association with diabetes was observed for either SUR1 variant. However, in contrast, analysis of the Kir6.2 E23K variant showed that the KK homozygosity was more frequent in Type 2 diabetic than control subjects. Variants were not associated with clinical characteristics nor did they affect response to sulphonylurea therapyThere is no support at present for mutations in either Kir6.2 or SUR1 promoter sequences contributing to Type 2 diabetes. However, the minimal promoter region of SUR1 has yet to be investigated. The E23K variant of Kir6.2 is associated with Type 2 diabetes mellitus in the UKPDS cohort.

    View details for DOI 10.1046/j.1464-5491.2001.00449.x

    View details for PubMedID 11318841