Honors & Awards


  • Reed-Hodgson Professor, Human Biology (2004-present)
  • Katherine Dexter McCormick and Stanley McCormick Memorial Professor, Stanford University School of Medicine (2022 - present)
  • Member, American Academy of Arts and Sciences (2006-present)
  • Member, National Academy of Sciences (2008-present)
  • Member, Institute of Medicine (2011-present)

Professional Education


  • Ph. D., Mass Inst Tech, Microbiology (1980)
  • B. A., Brandeis Univ., Physics (1974)
  • Postdoctoral, Indiana University, Developmental Genetics (1983)

Current Research and Scholarly Interests


My laboratory uses the Drosophila male germ line as a model to investigate how self-renewal, proliferation and differentiation are regulated in adult stem cell lineages. The central characteristic of adult stem cells is their long-term capacity to divide as relatively undifferentiated precursors while also producing daughter cells that initiate differentiation. Understanding the mechanisms that regulate stem cell specification and the choice between stem cell self-renewal and differentiation is crucial for realizing the potential of stem cells for regenerative medicine. We are using the Drosophila male germ line as a powerful genetic system to identify both the cell autonomous determinants and the extrinsic cell-cell interactions that govern stem cell specification, self-renewal, and differentiation. One of the great advantages of this system is that stem cells can be studied in situ, in the context of their normal support cells. Our results indicate that signals from surrounding somatic support cells specify asymmetric division of male germ line stem cells by inducing one daughter cell to self-renew stem cell identity while directing the other daughter cell to differentiate. A second focus of our work concerns how the developmental program directs cellular differentiation. Fundamental cellular functions like the cell cycle, the cytoskeleton, and the general transcription machinery are remodeled during development to give rise to specialized cell types. Several lines of research in our laboratory have recently converged on the molecular mechanisms underlying the developmentally programmed switch from proliferation to differentiation, a key regulatory point in the adult stem cell lineages that underlie tissue maintenance and repair. Failure to cleanly execute this switch may contribute to genesis of cancer. Our results implicate a number of molecular and cellular mechanisms in regulating this critical switch. We find that RNA binding proteins involved in translational control and alternative splicing act cell autonomously to regulate the cessation of proliferation and that progression of differentiation requires communication from associated somatic support cells. We discovered that a developmentally regulated alternate choice of site at which certain nascent transcripts are cut to form 3’ ends, leading to production of novel mRNA isoforms with shortened 3’UTRs, controls dramatic changes in the suite of proteins expressed in differentiating spermatocytes compared to proliferating spermatogonia. We found that dramatic changes in chromatin open over 2000 new promoters with novel core sequence structure to turn on the new cell type specific transcription program when cells initiate spermatocyte differentiation. Some of the earliest genes turned on in this differentiation program encode chromatin associated proteins that prevent spurious opening of normally cryptic promoters, thus preventing massive misexpression of genes associated with the wrong cell type. Other transcripts upregulated with differentiation onset encode cell type-specific translational regulators that delay production of core G2/M cell cycle machinery to program the extended G2 phase of meiotic prophase. Our goal over the next 5 years is to map how these processes collaborate to form the regulatory circuitry that initiates then executes the switch from proliferation to differentiation.

Clinical Trials


  • Testing the Safety and Tolerability of CX-4945 in Patients With Recurrent Medulloblastoma Who May or May Not Have Surgery Not Recruiting

    This is a multi center, Phase I, Phase II and surgical study of the CX-4945 drug (silmitasertib sodium) for patients with recurrent SHH (Sonic Hedgehog) medulloblastoma

    Stanford is currently not accepting patients for this trial. For more information, please contact Cancer Clinical Trials Office (CCTO), 650-498-7061.

    View full details

2024-25 Courses


Stanford Advisees


All Publications


  • A developmental mechanism to regulate alternative polyadenylation in an adult stem cell lineage. Genes & development Gallicchio, L., Matias, N. R., Morales-Polanco, F., Nava, I., Stern, S., Zeng, Y., Fuller, M. T. 2024

    Abstract

    Alternative cleavage and polyadenylation (APA) often results in production of mRNA isoforms with either longer or shorter 3' UTRs from the same genetic locus, potentially impacting mRNA translation, localization, and stability. Developmentally regulated APA can thus make major contributions to cell type-specific gene expression programs as cells differentiate. During Drosophila spermatogenesis, ∼500 genes undergo APA when proliferating spermatogonia differentiate into spermatocytes, producing transcripts with shortened 3' UTRs, leading to profound stage-specific changes in the proteins expressed. The molecular mechanisms that specify usage of upstream polyadenylation sites in spermatocytes are thus key to understanding the changes in cell state. Here, we show that upregulation of PCF11 and Cbc, the two components of cleavage factor II (CFII), orchestrates APA during Drosophila spermatogenesis. Knockdown of PCF11 or cbc in spermatocytes caused dysregulation of APA, with many transcripts normally cleaved at a proximal site in spermatocytes now cleaved at their distal site, as in spermatogonia. Forced overexpression of CFII components in spermatogonia switched cleavage of some transcripts to the proximal site normally used in spermatocytes. Our findings reveal a developmental mechanism where changes in expression of specific cleavage factors can direct cell type-specific APA at selected genes.

    View details for DOI 10.1101/gad.351649.124

    View details for PubMedID 39111825

  • Developmental Regulation of Alternative Polyadenylation in an Adult Stem Cell Lineage. bioRxiv : the preprint server for biology Gallicchio, L., R Matias, N., Morales-Polanco, F., Nava, I., Stern, S., Zeng, Y., Fuller, M. T. 2024

    Abstract

    Co-transcriptional alternate processing of nascent mRNA molecules can make major contributions to cell type specific gene expression programs as proliferating precursor cells initiate terminal differentiation. Alternative Cleavage and Polyadenylation (APA) can result in the production of mRNA isoforms from the same gene locus with either longer or shorter 3'UTRs. In Drosophila spermatogenesis, approximately 500 genes undergo APA as proliferating spermatogonia differentiate into spermatocytes, producing transcript isoforms with shortened 3'UTRs, and resulting in profound stage specific changes in the proteins expressed. The molecular mechanisms that specify usage of upstream polyadenylation sites in spermatocytes are thus key to understanding the changes in cell state. Here, we show that PCF11 and Cbc, the two components of Cleavage factor II (CFII), orchestrate APA switching during Drosophila spermatogenesis. Knockdown of PCF11 or cbc in spermatocytes caused dysregulation of APA, with many transcripts normally cleaved at a proximal site in spermatocytes now cleaved at their distal site, as in spermatogonia. Although PCF11 is widely expressed, cbc is strongly upregulated in spermatocytes. Our findings reveal a developmental mechanism where changes in activity of specific cleavage factors can direct cell type specific APA at selected genes, presenting CFII as a key developmental regulator of APA during spermatogenesis.

    View details for DOI 10.1101/2024.03.18.585561

    View details for PubMedID 38562704

  • Cell-type-specific interacting proteins collaborate to regulate the timing of Cyclin B protein expression in male meiotic prophase. Development (Cambridge, England) Baker, C. C., Gallicchio, L., Matias, N. R., Porter, D. F., Parsanian, L., Taing, E., Tam, C., Fuller, M. T. 2023

    Abstract

    During meiosis, germ cell and stage-specific components impose additional layers of regulation on the core cell cycle machinery to set up an extended G2 period termed meiotic prophase. In Drosophila males, meiotic prophase lasts 3.5 days, during which spermatocytes upregulate of over 1800 genes and grow 25-fold. Previous work showed that the cell cycle regulator Cyclin B (CycB) is subject to translational repression in immature spermatocytes, mediated by the RNA-binding protein Rbp4 and its partner Fest. Here we show that the spermatocyte-specific protein Lut is required for translational repression of cycB in an 8-hour window just before spermatocytes are fully mature. In males mutant for rbp4 or lut, spermatocytes enter and exit meiotic division 6-8 hours earlier than in wild-type. In addition, spermatocyte-specific isoforms of Syncrip (Syp) are required for expression of CycB protein in mature spermatocytes and normal entry into the meiotic divisions. Lut and Syp interact with Fest independent of RNA. Thus a set of spermatocyte-specific regulators choreograph the timing of expression of CycB protein during male meiotic prophase.

    View details for DOI 10.1242/dev.201709

    View details for PubMedID 37882771

  • Developmentally regulated alternate 3' end cleavage of nascent transcripts controls dynamic changes in protein expression in an adult stem cell lineage. Genes & development Berry, C. W., Olivares, G. H., Gallicchio, L., Ramaswami, G., Glavic, A., Olguín, P., Li, J. B., Fuller, M. T. 2022

    Abstract

    Alternative polyadenylation (APA) generates transcript isoforms that differ in the position of the 3' cleavage site, resulting in the production of mRNA isoforms with different length 3' UTRs. Although widespread, the role of APA in the biology of cells, tissues, and organisms has been controversial. We identified >500 Drosophila genes that express mRNA isoforms with a long 3' UTR in proliferating spermatogonia but a short 3' UTR in differentiating spermatocytes due to APA. We show that the stage-specific choice of the 3' end cleavage site can be regulated by the arrangement of a canonical polyadenylation signal (PAS) near the distal cleavage site but a variant or no recognizable PAS near the proximal cleavage site. The emergence of transcripts with shorter 3' UTRs in differentiating cells correlated with changes in expression of the encoded proteins, either from off in spermatogonia to on in spermatocytes or vice versa. Polysome gradient fractionation revealed >250 genes where the long 3' UTR versus short 3' UTR mRNA isoforms migrated differently, consistent with dramatic stage-specific changes in translation state. Thus, the developmentally regulated choice of an alternative site at which to make the 3' end cut that terminates nascent transcripts can profoundly affect the suite of proteins expressed as cells advance through sequential steps in a differentiation lineage.

    View details for DOI 10.1101/gad.349689.122

    View details for PubMedID 36175033

  • Fly Cell Atlas: A single-nucleus transcriptomic atlas of the adult fruit fly. Science (New York, N.Y.) Li, H., Janssens, J., De Waegeneer, M., Kolluru, S. S., Davie, K., Gardeux, V., Saelens, W., David, F. P., Brbic, M., Spanier, K., Leskovec, J., McLaughlin, C. N., Xie, Q., Jones, R. C., Brueckner, K., Shim, J., Tattikota, S. G., Schnorrer, F., Rust, K., Nystul, T. G., Carvalho-Santos, Z., Ribeiro, C., Pal, S., Mahadevaraju, S., Przytycka, T. M., Allen, A. M., Goodwin, S. F., Berry, C. W., Fuller, M. T., White-Cooper, H., Matunis, E. L., DiNardo, S., Galenza, A., O'Brien, L. E., Dow, J. A., FCA Consortium, Jasper, H., Oliver, B., Perrimon, N., Deplancke, B., Quake, S. R., Luo, L., Aerts, S., Agarwal, D., Ahmed-Braimah, Y., Arbeitman, M., Ariss, M. M., Augsburger, J., Ayush, K., Baker, C. C., Banisch, T., Birker, K., Bodmer, R., Bolival, B., Brantley, S. E., Brill, J. A., Brown, N. C., Buehner, N. A., Cai, X. T., Cardoso-Figueiredo, R., Casares, F., Chang, A., Clandinin, T. R., Crasta, S., Desplan, C., Detweiler, A. M., Dhakan, D. B., Dona, E., Engert, S., Floc'hlay, S., George, N., Gonzalez-Segarra, A. J., Groves, A. K., Gumbin, S., Guo, Y., Harris, D. E., Heifetz, Y., Holtz, S. L., Horns, F., Hudry, B., Hung, R., Jan, Y. N., Jaszczak, J. S., Jefferis, G. S., Karkanias, J., Karr, T. L., Katheder, N. S., Kezos, J., Kim, A. A., Kim, S. K., Kockel, L., Konstantinides, N., Kornberg, T. B., Krause, H. M., Labott, A. T., Laturney, M., Lehmann, R., Leinwand, S., Li, J., Li, J. S., Li, K., Li, K., Li, L., Li, T., Litovchenko, M., Liu, H., Liu, Y., Lu, T., Manning, J., Mase, A., Matera-Vatnick, M., Matias, N. R., McDonough-Goldstein, C. E., McGeever, A., McLachlan, A. D., Moreno-Roman, P., Neff, N., Neville, M., Ngo, S., Nielsen, T., O'Brien, C. E., Osumi-Sutherland, D., Ozel, M. N., Papatheodorou, I., Petkovic, M., Pilgrim, C., Pisco, A. O., Reisenman, C., Sanders, E. N., Dos Santos, G., Scott, K., Sherlekar, A., Shiu, P., Sims, D., Sit, R. V., Slaidina, M., Smith, H. E., Sterne, G., Su, Y., Sutton, D., Tamayo, M., Tan, M., Tastekin, I., Treiber, C., Vacek, D., Vogler, G., Waddell, S., Wang, W., Wilson, R. I., Wolfner, M. F., Wong, Y. E., Xie, A., Xu, J., Yamamoto, S., Yan, J., Yao, Z., Yoda, K., Zhu, R., Zinzen, R. P. 2022; 375 (6584): eabk2432

    Abstract

    For more than 100 years, the fruit fly Drosophila melanogaster has been one of the most studied model organisms. Here, we present a single-cell atlas of the adult fly, Tabula Drosophilae, that includes 580,000 nuclei from 15 individually dissected sexed tissues as well as the entire head and body, annotated to >250 distinct cell types. We provide an in-depth analysis of cell type-related gene signatures and transcription factor markers, as well as sexual dimorphism, across the whole animal. Analysis of common cell types between tissues, such as blood and muscle cells, reveals rare cell types and tissue-specific subtypes. This atlas provides a valuable resource for the Drosophila community and serves as a reference to study genetic perturbations and disease models at single-cell resolution.

    View details for DOI 10.1126/science.abk2432

    View details for PubMedID 35239393

  • Developmental regulation of cell type-specific transcription by novel promoter-proximal sequence elements. Genes & development Lu, D. n., Sin, H. S., Lu, C. n., Fuller, M. T. 2020

    Abstract

    Cell type-specific transcriptional programs that drive differentiation of specialized cell types are key players in development and tissue regeneration. One of the most dramatic changes in the transcription program in Drosophila occurs with the transition from proliferating spermatogonia to differentiating spermatocytes, with >3000 genes either newly expressed or expressed from new alternative promoters in spermatocytes. Here we show that opening of these promoters from their closed state in precursor cells requires function of the spermatocyte-specific tMAC complex, localized at the promoters. The spermatocyte-specific promoters lack the previously identified canonical core promoter elements except for the Inr. Instead, these promoters are enriched for the binding site for the TALE-class homeodomain transcription factors Achi/Vis and for a motif originally identified under tMAC ChIP-seq peaks. The tMAC motif resembles part of the previously identified 14-bp β2UE1 element critical for spermatocyte-specific expression. Analysis of downstream sequences relative to transcription start site usage suggested that ACA and CNAAATT motifs at specific positions can help promote efficient transcription initiation. Our results reveal how promoter-proximal sequence elements that recruit and are acted upon by cell type-specific chromatin binding complexes help establish a robust, cell type-specific transcription program for terminal differentiation.

    View details for DOI 10.1101/gad.335331.119

    View details for PubMedID 32217666

  • Somatic support cells regulate germ cell survival through the Baz/aPKC/Par6 complex DEVELOPMENT Brantley, S. E., Fuller, M. T. 2019; 146 (8)

    View details for DOI 10.1242/dev.169342

    View details for Web of Science ID 000467762400007

  • Developmental phosphoproteomics identifies the kinase CK2 as a driver of Hedgehog signaling and a therapeutic target in medulloblastoma. Science signaling Purzner, T., Purzner, J., Buckstaff, T., Cozza, G., Gholamin, S., Rusert, J. M., Hartl, T. A., Sanders, J., Conley, N., Ge, X., Langan, M., Ramaswamy, V., Ellis, L., Litzenburger, U., Bolin, S., Theruvath, J., Nitta, R., Qi, L., Li, X., Li, G., Taylor, M. D., Wechsler-Reya, R. J., Pinna, L. A., Cho, Y., Fuller, M. T., Elias, J. E., Scott, M. P. 2018; 11 (547)

    Abstract

    A major limitation of targeted cancer therapy is the rapid emergence of drug resistance, which often arises through mutations at or downstream of the drug target or through intrinsic resistance of subpopulations of tumor cells. Medulloblastoma (MB), the most common pediatric brain tumor, is no exception, and MBs that are driven by sonic hedgehog (SHH) signaling are particularly aggressive and drug-resistant. To find new drug targets and therapeutics for MB that may be less susceptible to common resistance mechanisms, we used a developmental phosphoproteomics approach in murine granule neuron precursors (GNPs), the developmental cell of origin of MB. The protein kinase CK2 emerged as a driver of hundreds of phosphorylation events during the proliferative, MB-like stage of GNP growth, including the phosphorylation of three of the eight proteins commonly amplified in MB. CK2 was critical to the stabilization and activity of the transcription factor GLI2, a late downstream effector in SHH signaling. CK2 inhibitors decreased the viability of primary SHH-type MB patient cells in culture and blocked the growth of murine MB tumors that were resistant to currently available Hh inhibitors, thereby extending the survival of tumor-bearing mice. Because of structural interactions, one CK2 inhibitor (CX-4945) inhibited both wild-type and mutant CK2, indicating that this drug may avoid at least one common mode of acquired resistance. These findings suggest that CK2 inhibitors may be effective for treating patients with MB and show how phosphoproteomics may be used to gain insight into developmental biology and pathology.

    View details for PubMedID 30206138

  • The conserved RNA helicase YTHDC2 regulates the transition from proliferation to differentiation in the germline ELIFE Bailey, A. S., Batista, P. J., Gold, R. S., Chen, Y., de Rooij, D. G., Chang, H. Y., Fuller, M. T. 2017; 6

    Abstract

    The switch from mitosis to meiosis is the key event marking onset of differentiation in the germline stem cell lineage. In Drosophila, the translational repressor Bgcn is required for spermatogonia to stop mitosis and transition to meiotic prophase and the spermatocyte state. Here we show that the mammalian Bgcn homolog YTHDC2 facilitates a clean switch from mitosis to meiosis in mouse germ cells, revealing a conserved role for YTHDC2 in this critical cell fate transition. YTHDC2-deficient male germ cells enter meiosis but have a mixed identity, maintaining expression of Cyclin A2 and failing to properly express many meiotic markers. Instead of continuing through meiotic prophase, the cells attempt an abnormal mitotic-like division and die. YTHDC2 binds multiple transcripts including Ccna2 and other mitotic transcripts, binds specific piRNA precursors, and interacts with RNA granule components, suggesting that proper progression of germ cells through meiosis is licensed by YTHDC2 through post-transcriptional regulation.

    View details for PubMedID 29087293

  • Blocking promiscuous activation at cryptic promoters directs cell type-specific gene expression SCIENCE Kim, J., Lu, C., Srinivasan, S., Awe, S., Brehm, A., Fuller, M. T. 2017; 356 (6339): 717-721

    Abstract

    To selectively express cell type-specific transcripts during development, it is critical to maintain genes required for other lineages in a silent state. Here, we show in the Drosophila male germline stem cell lineage that a spermatocyte-specific zinc finger protein, Kumgang (Kmg), working with the chromatin remodeler dMi-2 prevents transcription of genes normally expressed only in somatic lineages. By blocking transcription from normally cryptic promoters, Kmg restricts activation by Aly, a component of the testis-meiotic arrest complex, to transcripts for male germ cell differentiation. Our results suggest that as new regions of the genome become open for transcription during terminal differentiation, blocking the action of a promiscuous activator on cryptic promoters is a critical mechanism for specifying precise gene activation.

    View details for DOI 10.1126/science.aal3096

    View details for Web of Science ID 000401508400039

    View details for PubMedID 28522526

  • Cell type-specific translational repression of Cyclin B during meiosis in males DEVELOPMENT Baker, C. C., Gim, B. S., Fuller, M. T. 2015; 142 (19): 3394-3402

    Abstract

    The unique cell cycle dynamics of meiosis are controlled by layers of regulation imposed on core mitotic cell cycle machinery components by the program of germ cell development. Although the mechanisms that regulate Cdk1/Cyclin B activity in meiosis in oocytes have been well studied, little is known about the trans-acting factors responsible for developmental control of these factors in male gametogenesis. During meiotic prophase in Drosophila males, transcript for the core cell cycle protein Cyclin B1 (CycB) is expressed in spermatocytes, but the protein does not accumulate in spermatocytes until just before the meiotic divisions. Here, we show that two interacting proteins, Rbp4 and Fest, expressed at the onset of spermatocyte differentiation under control of the developmental program of male gametogenesis, function to direct cell type- and stage-specific repression of translation of the core G2/M cell cycle component cycB during the specialized cell cycle of male meiosis. Binding of Fest to Rbp4 requires a 31-amino acid region within Rbp4. Rbp4 and Fest are required for translational repression of cycB in immature spermatocytes, with Rbp4 binding sequences in a cell type-specific shortened form of the cycB 3' UTR. Finally, we show that Fest is required for proper execution of meiosis I.

    View details for DOI 10.1242/dev.122341

    View details for Web of Science ID 000365025500014

    View details for PubMedID 26443637

    View details for PubMedCentralID PMC4631753

  • Somatic cell lineage is required for differentiation and not maintenance of germline stem cells in Drosophila testes PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Lim, J. G., Fuller, M. T. 2012; 109 (45): 18477-18481

    Abstract

    Adult stem cells are believed to be maintained by a specialized microenvironment, the niche, which provides short-range signals that either instruct stem cells to self-renew or inhibit execution of preprogrammed differentiation pathways. In Drosophila testes, somatic cyst stem cells (CySCs) and the apical hub form the niche for neighboring germline stem cells (GSCs), with CySCs as the proposed source of instructive self-renewal signals [Leatherman JL, Dinardo S (2010) Nat Cell Biol 12(8):806-811]. In contrast to this model, we show that early germ cells with GSC characteristics can be maintained over time after ablation of CySCs and their cyst cell progeny. Without CySCs and cyst cells, early germ cells away from the hub failed to initiate differentiation. Our results suggest that CySCs do not have a necessary instructive role in specifying GSC self-renewal and that the differentiated progeny of CySCs provide an environment necessary to trigger GSC differentiation. This work highlights the complex interaction between different stem cell populations in the same niche and how the state of one stem cell population can influence the fate of the other.

    View details for DOI 10.1073/pnas.1215516109

    View details for Web of Science ID 000311156700055

    View details for PubMedID 23091022

    View details for PubMedCentralID PMC3494938

  • A Self-Limiting Switch Based on Translational Control Regulates the Transition from Proliferation to Differentiation in an Adult Stem Cell Lineage CELL STEM CELL Insco, M. L., Bailey, A. S., Kim, J., Olivares, G. H., Wapinski, O. L., Tam, C. H., Fuller, M. T. 2012; 11 (5): 689-700

    Abstract

    In adult stem cell lineages, progenitor cells commonly undergo mitotic transit amplifying (TA) divisions before terminal differentiation, allowing production of many differentiated progeny per stem cell division. Mechanisms that limit TA divisions and trigger the switch to differentiation may protect against cancer by preventing accumulation of oncogenic mutations in the proliferating population. Here we show that the switch from TA proliferation to differentiation in the Drosophila male germline stem cell lineage is mediated by translational control. The TRIM-NHL tumor suppressor homolog Mei-P26 facilitates accumulation of the differentiation regulator Bam in TA cells. In turn, Bam and its partner Bgcn bind the mei-P26 3' untranslated region and repress translation of mei-P26 in late TA cells. Thus, germ cells progress through distinct, sequential regulatory states, from Mei-P26 on/Bam off to Bam on/Mei-P26 off. TRIM-NHL homologs across species facilitate the switch from proliferation to differentiation, suggesting a conserved developmentally programmed tumor suppressor mechanism.

    View details for DOI 10.1016/j.stem.2012.08.012

    View details for Web of Science ID 000311471900014

    View details for PubMedID 23122292

  • Asymmetric inheritance of mother versus daughter centrosome in stem cell division SCIENCE Yamashita, Y. M., Mahowald, A. P., Perlin, J. R., Fuller, M. T. 2007; 315 (5811): 518-521

    Abstract

    Adult stem cells often divide asymmetrically to produce one self-renewed stem cell and one differentiating cell, thus maintaining both populations. The asymmetric outcome of stem cell divisions can be specified by an oriented spindle and local self-renewal signals from the stem cell niche. Here we show that developmentally programmed asymmetric behavior and inheritance of mother and daughter centrosomes underlies the stereotyped spindle orientation and asymmetric outcome of stem cell divisions in the Drosophila male germ line. The mother centrosome remains anchored near the niche while the daughter centrosome migrates to the opposite side of the cell before spindle formation.

    View details for DOI 10.1126/science.1134910

    View details for Web of Science ID 000243726600048

    View details for PubMedID 17255513

    View details for PubMedCentralID PMC2563045

  • Tissue-specific TAFs counteract polycomb to turn on terminal differentiation SCIENCE Chen, X., Hiller, M., Sancak, Y., Fuller, M. T. 2005; 310 (5749): 869-872

    Abstract

    Polycomb transcriptional silencing machinery is implicated in the maintenance of precursor fates, but how this repression is reversed to allow cell differentiation is unknown. Here we show that testis-specific TAF (TBP-associated factor) homologs required for terminal differentiation of male germ cells may activate target gene expression in part by counteracting repression by Polycomb. Chromatin immunoprecipitation revealed that testis TAFs bind to target promoters, reduce Polycomb binding, and promote local accumulation of H3K4me3, a mark of Trithorax action. Testis TAFs also promoted relocalization of Polycomb Repression Complex 1 components to the nucleolus in spermatocytes, implicating subnuclear architecture in the regulation of terminal differentiation.

    View details for DOI 10.1126/science.1118101

    View details for Web of Science ID 000233121800050

    View details for PubMedID 16272126

  • Orientation of asymmetric stem cell division by the APC tumor suppressor and centrosome SCIENCE Yamashita, Y. M., Jones, D. L., Fuller, M. T. 2003; 301 (5639): 1547-1550

    Abstract

    Stem cell self-renewal can be specified by local signals from the surrounding microenvironment, or niche. However, the relation between the niche and the mechanisms that ensure the correct balance between stem cell self-renewal and differentiation is poorly understood. Here, we show that dividing Drosophila male germline stem cells use intracellular mechanisms involving centrosome function and cortically localized Adenomatous Polyposis Coli tumor suppressor protein to orient mitotic spindles perpendicular to the niche, ensuring a reliably asymmetric outcome in which one daughter cell remains in the niche and self-renews stem cell identity, whereas the other, displaced away, initiates differentiation.

    View details for Web of Science ID 000185255300054

    View details for PubMedID 12970569

  • Functional septate junctions between cyst cells are required for survival of transit amplifying male germ cells expressing Bag of marbles. bioRxiv : the preprint server for biology Berry, C. W., Fuller, M. T. 2024

    Abstract

    In adult stem cell lineages, the cellular microenvironment plays essential roles to ensure the proper balance of self-renewal, differentiation and regulated elimination of differentiating cells. Although regulated death of progenitor cells undergoing proliferation or early differentiation is a feature of many tissues, mechanisms that initiate this pruning remain unexplored, particularly in the male germline, where up to 30% of the germline is eliminated before the meiotic divisions. We conducted a targeted screen to identify functional regulators required in somatic support cells for survival or differentiation at early steps in the male germ line stem cell lineage. Cell type-specific knockdown in cyst cells uncovered novel roles of genes in germline stem cell differentiation, including a previously unappreciated role of the Septate Junction (SJ) in preventing cell death of differentiating germline progenitors. Loss of the SJ in the somatic cyst cells resulted in elimination of transit-amplifying spermatogonia by the 8-cell stage. Germ cell death was spared in males mutant for the differentiation factor bam indicating that intact barriers surrounding transit amplifying progenitors are required to ensure germline survival once differentiation has initiated.

    View details for DOI 10.1101/2024.04.02.587826

    View details for PubMedID 38617328

  • An organism-wide atlas of hormonal signaling based on the mouse lemur single-cell transcriptome. Nature communications Liu, S., Ezran, C., Wang, M. F., Li, Z., Awayan, K., Long, J. Z., De Vlaminck, I., Wang, S., Epelbaum, J., Kuo, C. S., Terrien, J., Krasnow, M. A., Ferrell, J. E. 2024; 15 (1): 2188

    Abstract

    Hormones mediate long-range cell communication and play vital roles in physiology, metabolism, and health. Traditionally, endocrinologists have focused on one hormone or organ system at a time. Yet, hormone signaling by its very nature connects cells of different organs and involves crosstalk of different hormones. Here, we leverage the organism-wide single cell transcriptional atlas of a non-human primate, the mouse lemur (Microcebus murinus), to systematically map source and target cells for 84 classes of hormones. This work uncovers previously-uncharacterized sites of hormone regulation, and shows that the hormonal signaling network is densely connected, decentralized, and rich in feedback loops. Evolutionary comparisons of hormonal genes and their expression patterns show that mouse lemur better models human hormonal signaling than mouse, at both the genomic and transcriptomic levels, and reveal primate-specific rewiring of hormone-producing/target cells. This work complements the scale and resolution of classical endocrine studies and sheds light on primate hormone regulation.

    View details for DOI 10.1038/s41467-024-46070-9

    View details for PubMedID 38467625

    View details for PubMedCentralID 1540572

  • Emergent dynamics of adult stem cell lineages from single nucleus and single cell RNA-Seq of Drosophila testes. eLife Raz, A. A., Vida, G. S., Stern, S. R., Mahadevaraju, S., Fingerhut, J. M., Viveiros, J. M., Pal, S., Grey, J. R., Grace, M. R., Berry, C. W., Li, H., Janssens, J., Saelens, W., Shao, Z., Hu, C., Yamashita, Y. M., Przytycka, T., Oliver, B., Brill, J. A., Krause, H., Matunis, E. L., White-Cooper, H., DiNardo, S., Fuller, M. T. 2023; 12

    Abstract

    Proper differentiation of sperm from germline stem cells, essential for production of the next generation, requires dramatic changes in gene expression that drive remodeling of almost all cellular components, from chromatin to organelles to cell shape itself. Here, we provide a single nucleus and single cell RNA-seq resource covering all of spermatogenesis in Drosophila starting from in-depth analysis of adult testis single nucleus RNA-seq (snRNA-seq) data from the Fly Cell Atlas (FCA) study. With over 44,000 nuclei and 6000 cells analyzed, the data provide identification of rare cell types, mapping of intermediate steps in differentiation, and the potential to identify new factors impacting fertility or controlling differentiation of germline and supporting somatic cells. We justify assignment of key germline and somatic cell types using combinations of known markers, in situ hybridization, and analysis of extant protein traps. Comparison of single cell and single nucleus datasets proved particularly revealing of dynamic developmental transitions in germline differentiation. To complement the web-based portals for data analysis hosted by the FCA, we provide datasets compatible with commonly used software such as Seurat and Monocle. The foundation provided here will enable communities studying spermatogenesis to interrogate the datasets to identify candidate genes to test for function in vivo.

    View details for DOI 10.7554/eLife.82201

    View details for PubMedID 36795469

  • YTHDC2 serves a distinct late role in spermatocytes during germ cell differentiation. bioRxiv : the preprint server for biology Bailey, A. S., Fuller, M. T. 2023

    Abstract

    Post-transcriptional regulation of gene expression by RNA-binding proteins helps facilitate fast, clean transitions from one cell state to the next during germ cell differentiation. Previously we showed that the RNA helicase YTHDC2 is required for germ cells to properly switch from mitosis to meiosis (Bailey et al., 2017). While YTHDC2 protein is first expressed as male germ cells enter meiosis, when it is needed to shut down the mitotic program, YTHDC2 expression continues to increase and reaches its highest levels later in meiotic prophase, in pachytene spermatocytes. Here we show that YTHDC2 has an additional essential role regulating meiotic progression in late spermatocytes during mouse germ cell differentiation. Inducing conditional knockout of Ythdc2 during the first wave of spermatogenesis, after the germ cells have already initiated meiotic prophase, allowed Ythdc2 -deficient germ cells to successfully reach the pachytene stage and properly express many meiotic markers. However, instead of continuing through meiotic prophase and initiating the meiotic divisions, late pachytene spermatocytes failed to transition to the diplotene stage and quickly died. Loss of function of Ythdc2 in spermatocytes resulted in changes in transcript levels for a number of genes, some up-regulated and some down-regulated, compared to control mid-stage spermatocytes. YTHDC2 interacts with different proteins in early and late spermatocytes, with many of the interacting proteins involved in post-transcriptional RNA regulation and present in RNA granules, similar to YTHDC2. Our findings suggest that YTHDC2 facilitates proper progression of germ cells through multiple steps of meiosis, potentially via several mechanisms of post-transcriptional RNA regulation.

    View details for DOI 10.1101/2023.01.23.525146

    View details for PubMedID 36747642

  • Regulation and function of alternative polyadenylation in development and differentiation. RNA biology Gallicchio, L., Olivares, G. H., Berry, C. W., Fuller, M. T. 2023; 20 (1): 908-925

    Abstract

    Alternative processing of nascent mRNAs is widespread in eukaryotic organisms and greatly impacts the output of gene expression. Specifically, alternative cleavage and polyadenylation (APA) is a co-transcriptional molecular process that switches the polyadenylation site (PAS) at which a nascent mRNA is cleaved, resulting in mRNA isoforms with different 3'UTR length and content. APA can potentially affect mRNA translation efficiency, localization, stability, and mRNA seeded protein-protein interactions. APA naturally occurs during development and cellular differentiation, with around 70% of human genes displaying APA in particular tissues and cell types. For example, neurons tend to express mRNAs with long 3'UTRs due to preferential processing at PASs more distal than other PASs used in other cell types. In addition, changes in APA mark a variety of pathological states, including many types of cancer, in which mRNAs are preferentially cleaved at more proximal PASs, causing expression of mRNA isoforms with short 3'UTRs. Although APA has been widely reported, both the function of APA in development and the mechanisms that regulate the choice of 3'end cut sites in normal and pathogenic conditions are still poorly understood. In this review, we summarize current understanding of how APA is regulated during development and cellular differentiation and how the resulting change in 3'UTR content affects multiple aspects of gene expression. With APA being a widespread phenomenon, the advent of cutting-edge scientific techniques and the pressing need for in-vivo studies, there has never been a better time to delve into the intricate mechanisms of alternative cleavage and polyadenylation.

    View details for DOI 10.1080/15476286.2023.2275109

    View details for PubMedID 37906624

  • Identification of Protein-RNA Interactions in Mouse Testis Tissue Using fRIP BIO-PROTOCOL Bailey, A., Batista, P., Chang, H., Fuller, M. 2022; 12 (01)
  • Identification of Protein-RNA Interactions in Mouse Testis Tissue Using fRIP. Bio-protocol Bailey, A. S., Batista, P. J., Chang, H. Y., Fuller, M. T. 2022; 12 (1): e4286

    Abstract

    During development, cells must quickly switch from one cell state to the next to execute precise and timely differentiation. One method to ensure fast transitions in cell states is by controlling gene expression at the post-transcriptional level through action of RNA-binding proteins on mRNAs. The ability to accurately identify the RNA targets of RNA-binding proteins at specific stages is key to understanding the functional role of RNA-binding proteins during development. Here we describe an adapted formaldehyde RNA immunoprecipitation (fRIP) protocol to identify the in vivo RNA targets of a cytoplasmic RNA-binding protein, YTHDC2, from testis, during the first wave of spermatogenesis, at the stage when germ cells are shutting off the proliferative program and initiating terminal differentiation ( Bailey et al., 2017 ). This protocol enables quick and efficient identification of endogenous RNAs bound to an RNA-binding protein, and facilitates the monitoring of stage-specific changes during development.

    View details for DOI 10.21769/BioProtoc.4286

    View details for PubMedID 35118177

    View details for PubMedCentralID PMC8769761

  • DREF Genetically Counteracts Mi-2 and Caf1 to Regulate Adult Stem Cell Maintenance. PLoS genetics Angulo, B., Srinivasan, S., Bolival, B. J., Olivares, G. H., Spence, A. C., Fuller, M. T. 2019; 15 (6): e1008187

    Abstract

    Active adult stem cells maintain a bipotential state with progeny able to either self-renew or initiate differentiation depending on extrinsic signals from the surrounding microenvironment. However, the intrinsic gene regulatory networks and chromatin states that allow adult stem cells to make these cell fate choices are not entirely understood. Here we show that the transcription factor DNA Replication-related Element Factor (DREF) regulates adult stem cell maintenance in the Drosophila male germline. A temperature-sensitive allele of DREF described in this study genetically separated a role for DREF in germline stem cell self-renewal from the general roles of DREF in cell proliferation. The DREF temperature-sensitive allele caused defects in germline stem cell self-renewal but allowed viability and division of germline stem cells as well as cell viability, growth and division of somatic cyst stem cells in the testes and cells in the Drosophila eye. Germline stem cells mutant for the temperature sensitive DREF allele exhibited lower activation of a TGF-beta reporter, and their progeny turned on expression of the differentiation factor Bam prematurely. Results of genetic interaction analyses revealed that Mi-2 and Caf1/p55, components of the Nucleosome Remodeling and Deacetylase (NuRD) complex, genetically antagonize the role of DREF in germline stem cell maintenance. Taken together, these data suggest that DREF contributes to intrinsic components of the germline stem cell regulatory network that maintains competence to self-renew.

    View details for DOI 10.1371/journal.pgen.1008187

    View details for PubMedID 31226128

  • The Dig Module and Clathrin-Mediated Endocytosis Regulate EGFR Signaling and Cyst Cell-Germline Coordination in the Drosophila Testis STEM CELL REPORTS Papagiannouli, F., Berry, C., Fuller, M. T. 2019; 12 (5): 1024–40
  • The Dlg Module and Clathrin-Mediated Endocytosis Regulate EGFR Signaling and Cyst Cell-Germline Coordination in the Drosophila Testis. Stem cell reports Papagiannouli, F., Berry, C. W., Fuller, M. T. 2019

    Abstract

    Tissue homeostasis and repair relies on proper communication of stem cells and their differentiating daughters with the local tissue microenvironment. In the Drosophila male germline adult stem cell lineage, germ cells proliferate and progressively differentiate enclosed in supportive somatic cyst cells, forming a small organoid, the functional unit of differentiation. Here we show that cell polarity and vesicle trafficking influence signal transduction in cyst cells, with profound effects on the germ cells they enclose. Our data suggest that the cortical components Dlg, Scrib, Lgl and the clathrin-mediated endocytic (CME) machinery downregulate epidermal growth factor receptor (EGFR) signaling. Knockdown of dlg, scrib, lgl, or CME components in cyst cells resulted in germ cell death, similar to increased signal transduction via the EGFR, while lowering EGFR or downstream signaling components rescued the defects. This work provides insights into how cell polarity and endocytosis cooperate to regulate signal transduction and sculpt developing tissues.

    View details for PubMedID 31006632

  • Somatic support cells regulate germ cell survival through the Baz/aPKC/Par6 complex. Development (Cambridge, England) Brantley, S. E., Fuller, M. T. 2019

    Abstract

    Local signals and structural support from the surrounding cellular microenvironment play key roles in directing development in both embryonic organs and adult tissues. In Drosophila, male germ cells are intimately associated and co-differentiate with supporting somatic cells. Here we show that function of the Baz/aPKC/Par6 apical polarity complex in somatic cyst cells is required stage-specifically for survival of the germ cells they enclose. Although spermatogonia enclosed by cyst cells in which function of the Par-complex had been knocked down survive and proliferate, newly formed spermatocytes enclosed by cyst cells lacking Par complex proteins died soon after onset of meiotic prophase. Loss of Par complex function resulted in stage specific overactivation of the Jun-kinase (JNK) pathway in cyst cells. Knocking down expression of JNK pathway components or the GTPase Rab35 in cyst cells lacking Par complex function rescued survival of neighboring spermatocytes, suggesting that action of the apical polarity complex ensures germ cell survival by preventing JNK pathway activation, and that the mechanism by which cyst cells lacking Par complex function kill neighboring spermatocytes requires intracellular trafficking in somatic cyst cells.

    View details for PubMedID 30918053

  • Drosophila doublefault protein coordinates multiple events during male meiosis by controlling mRNA translation. Development (Cambridge, England) Sechi, S. n., Frappaolo, A. n., Karimpour-Ghahnavieh, A. n., Gottardo, M. n., Burla, R. n., Di Francesco, L. n., Szafer-Glusman, E. n., Schininà, E. n., Fuller, M. T., Saggio, I. n., Riparbelli, M. G., Callaini, G. n., Giansanti, M. G. 2019

    Abstract

    During the extended prophase of Drosophila gametogenesis, spermatocytes undergo robust gene transcription and store many transcripts in the cytoplasm in a repressed state, until translational activation of select mRNAs in later steps of spermatogenesis. Here we characterize the Drosophila Doublefault (Dbf) protein as a C2H2-zinc finger protein, primarily expressed in testes, that is required for normal meiotic division and spermiogenesis. Loss of Dbf causes premature centriole disengagement and affects spindle structure, chromosome segregation and cytokinesis. We show that Dbf interacts with the RNA binding protein Syncrip/hnRNPQ, a key regulator of localized translation in Drosophila We propose that the pleiotropic effects of dbf loss-of-function mutants are associated with the requirement for dbf function for translation of specific transcripts in spermatocytes. In agreement with this hypothesis, Dbf protein binds cyclin B mRNA and is essential for translation of cyclin B in mature spermatocytes.

    View details for DOI 10.1242/dev.183053

    View details for PubMedID 31645358

  • Testis-specific ATP synthase peripheral stalk subunits required for tissue-specific mitochondrial morphogenesis in Drosophila. BMC cell biology Sawyer, E. M., Brunner, E. C., Hwang, Y., Ivey, L. E., Brown, O., Bannon, M., Akrobetu, D., Sheaffer, K. E., Morgan, O., Field, C. O., Suresh, N., Gordon, M. G., Gunnell, E. T., Regruto, L. A., Wood, C. G., Fuller, M. T., Hales, K. G. 2017; 18 (1): 16

    Abstract

    In Drosophila early post-meiotic spermatids, mitochondria undergo dramatic shaping into the Nebenkern, a spherical body with complex internal structure that contains two interwrapped giant mitochondrial derivatives. The purpose of this study was to elucidate genetic and molecular mechanisms underlying the shaping of this structure.The knotted onions (knon) gene encodes an unconventionally large testis-specific paralog of ATP synthase subunit d and is required for internal structure of the Nebenkern as well as its subsequent disassembly and elongation. Knon localizes to spermatid mitochondria and, when exogenously expressed in flight muscle, alters the ratio of ATP synthase complex dimers to monomers. By RNAi knockdown we uncovered mitochondrial shaping roles for other testis-expressed ATP synthase subunits.We demonstrate the first known instance of a tissue-specific ATP synthase subunit affecting tissue-specific mitochondrial morphogenesis. Since ATP synthase dimerization is known to affect the degree of inner mitochondrial membrane curvature in other systems, the effect of Knon and other testis-specific paralogs of ATP synthase subunits may be to mediate differential membrane curvature within the Nebenkern.

    View details for DOI 10.1186/s12860-017-0132-1

    View details for PubMedID 28335714

    View details for PubMedCentralID PMC5364652

  • Testis-specific ATP synthase peripheral stalk subunits required for tissue-specific mitochondrial morphogenesis in Drosophila BMC CELL BIOLOGY Sawyer, E. M., Brunner, E. C., Hwang, Y., Ivey, L. E., Brown, O., Bannon, M., Akrobetu, D., Sheaffer, K. E., Morgan, O., Field, C. O., Suresh, N., Gordon, M. G., Gunnell, E. T., Regruto, L. A., Wood, C. G., Fuller, M. T., Hales, K. G. 2016; 18

    Abstract

    In Drosophila early post-meiotic spermatids, mitochondria undergo dramatic shaping into the Nebenkern, a spherical body with complex internal structure that contains two interwrapped giant mitochondrial derivatives. The purpose of this study was to elucidate genetic and molecular mechanisms underlying the shaping of this structure.The knotted onions (knon) gene encodes an unconventionally large testis-specific paralog of ATP synthase subunit d and is required for internal structure of the Nebenkern as well as its subsequent disassembly and elongation. Knon localizes to spermatid mitochondria and, when exogenously expressed in flight muscle, alters the ratio of ATP synthase complex dimers to monomers. By RNAi knockdown we uncovered mitochondrial shaping roles for other testis-expressed ATP synthase subunits.We demonstrate the first known instance of a tissue-specific ATP synthase subunit affecting tissue-specific mitochondrial morphogenesis. Since ATP synthase dimerization is known to affect the degree of inner mitochondrial membrane curvature in other systems, the effect of Knon and other testis-specific paralogs of ATP synthase subunits may be to mediate differential membrane curvature within the Nebenkern.

    View details for DOI 10.1186/s12860-017-0132-1

    View details for Web of Science ID 000397328600001

    View details for PubMedCentralID PMC5364652

  • Differentiation in Stem Cell Lineages and in Life: Explorations in the Male Germ Line Stem Cell Lineage ESSAYS ON DEVELOPMENTAL BIOLOGY, PT A Fuller, M. T. 2016; 116: 375-390

    Abstract

    I have been privileged to work on cellular differentiation during a great surge of discovery that has revealed the molecular mechanisms and genetic regulatory circuitry that control embryonic development and adult tissue maintenance and repair. Studying the regulation of proliferation and differentiation in the male germ line stem cell lineage has allowed us investigate how the developmental program imposes layers of additional controls on fundamental cellular processes like cell cycle progression and gene expression to give rise to the huge variety of specialized cell types in our bodies. We are beginning to understand how local signals from somatic support cells specify self-renewal versus differentiation in the stem cell niche at the apical tip of the testis. We are discovering the molecular events that block cell proliferation and initiate terminal differentiation at the switch from mitosis to meiosis-a signature event of the germ cell program. Our work is beginning to reveal how the developmental program that sets up the dramatic new cell type-specific transcription program that prepares germ cells for meiotic division and spermatid differentiation is turned on when cells become spermatocytes. I have had the privilege of working with incredible students, postdocs, and colleagues who have discovered, brainstormed, challenged, and refined our science and our ideas of how developmental pathways and cellular mechanisms work together to drive differentiation.

    View details for DOI 10.1016/bs.ctdb.2015.11.041

    View details for Web of Science ID 000376939100021

    View details for PubMedID 26970629

  • Recruitment of Mediator Complex by Cell Type and Stage-Specific Factors Required for Tissue-Specific TAF Dependent Gene Activation in an Adult Stem Cell Lineage PLOS GENETICS Lu, C., Fuller, M. T. 2015; 11 (12)
  • Recruitment of Mediator Complex by Cell Type and Stage-Specific Factors Required for Tissue-Specific TAF Dependent Gene Activation in an Adult Stem Cell Lineage. PLoS genetics Lu, C., Fuller, M. T. 2015; 11 (12): e1005701

    Abstract

    Onset of terminal differentiation in adult stem cell lineages is commonly marked by robust activation of new transcriptional programs required to make the appropriate differentiated cell type(s). In the Drosophila male germ line stem cell lineage, the switch from proliferating spermatogonia to spermatocyte is accompanied by one of the most dramatic transcriptional changes in the fly, as over 1000 new transcripts turn on in preparation for meiosis and spermatid differentiation. Here we show that function of the coactivator complex Mediator is required for activation of hundreds of new transcripts in the spermatocyte program. Mediator appears to act in a sequential hierarchy, with the testis activating Complex (tMAC), a cell type specific form of the Mip/dREAM general repressor, required to recruit Mediator subunits to the chromatin, and Mediator function required to recruit the testis TAFs (tTAFs), spermatocyte specific homologs of subunits of TFIID. Mediator, tMAC and the tTAFs co-regulate expression of a major set of spermatid differentiation genes. The Mediator subunit Med22 binds the tMAC component Topi when the two are coexpressed in S2 cells, suggesting direct recruitment. Loss of Med22 function in spermatocytes causes meiosis I maturation arrest male infertility, similar to loss of function of the tMAC subunits or the tTAFs. Our results illuminate how cell type specific versions of the Mip/dREAM complex and the general transcription machinery cooperate to drive selective gene activation during differentiation in stem cell lineages.

    View details for DOI 10.1371/journal.pgen.1005701

    View details for PubMedID 26624996

    View details for PubMedCentralID PMC4666660

  • Exocyst-Dependent Membrane Addition Is Required for Anaphase Cell Elongation and Cytokinesis in Drosophila PLOS GENETICS Giansanti, M. G., Vanderleest, T. E., Jewett, C. E., Sechi, S., Frappaolo, A., Fabian, L., Robinett, C. C., Brill, J. A., Loerke, D., Fuller, M. T., Blankenship, J. T. 2015; 11 (11)

    View details for DOI 10.1371/journal.pgen.1005632

    View details for PubMedID 26528720

  • Escargot Restricts Niche Cell to Stem Cell Conversion in the Drosophila Testis CELL REPORTS Voog, J., Sandall, S. L., Hime, G. R., Resende, L. P., Loza-Coll, M., Aslanian, A., Yates, J. R., Hunter, T., Fuller, M. T., Jones, L. 2014; 7 (3): 722-734

    Abstract

    Stem cells reside within specialized microenvironments, or niches, that control many aspects of stem cell behavior. Somatic hub cells in the Drosophila testis regulate the behavior of cyst stem cells (CySCs) and germline stem cells (GSCs) and are a primary component of the testis stem cell niche. The shutoff (shof) mutation, characterized by premature loss of GSCs and CySCs, was mapped to a locus encoding the evolutionarily conserved transcription factor Escargot (Esg). Hub cells depleted of Esg acquire CySC characteristics and differentiate as cyst cells, resulting in complete loss of hub cells and eventually CySCs and GSCs, similar to the shof mutant phenotype. We identified Esg-interacting proteins and demonstrate an interaction between Esg and the corepressor C-terminal binding protein (CtBP), which was also required for maintenance of hub cell fate. Our results indicate that niche cells can acquire stem cell properties upon removal of a single transcription factor in vivo.

    View details for DOI 10.1016/j.celrep.2014.04.025

    View details for Web of Science ID 000335560900015

    View details for PubMedID 24794442

    View details for PubMedCentralID PMC4128242

  • GOLPH3 Is Essential for Contractile Ring Formation and Rab11 Localization to the Cleavage Site during Cytokinesis in Drosophila melanogaster PLOS GENETICS Sechi, S., Colotti, G., Belloni, G., Mattei, V., Frappaolo, A., Raffa, G. D., Fuller, M. T., Giansanti, M. G. 2014; 10 (5)

    Abstract

    The highly conserved Golgi phosphoprotein 3 (GOLPH3) protein has been described as a Phosphatidylinositol 4-phosphate [PI(4)P] effector at the Golgi. GOLPH3 is also known as a potent oncogene, commonly amplified in several human tumors. However, the molecular pathways through which the oncoprotein GOLPH3 acts in malignant transformation are largely unknown. GOLPH3 has never been involved in cytokinesis. Here, we characterize the Drosophila melanogaster homologue of human GOLPH3 during cell division. We show that GOLPH3 accumulates at the cleavage furrow and is required for successful cytokinesis in Drosophila spermatocytes and larval neuroblasts. In premeiotic spermatocytes GOLPH3 protein is required for maintaining the organization of Golgi stacks. In dividing spermatocytes GOLPH3 is essential for both contractile ring and central spindle formation during cytokinesis. Wild type function of GOLPH3 enables maintenance of centralspindlin and Rho1 at cell equator and stabilization of Myosin II and Septin rings. We demonstrate that the molecular mechanism underlying GOLPH3 function in cytokinesis is strictly dependent on the ability of this protein to interact with PI(4)P. Mutations that abolish PI(4)P binding impair recruitment of GOLPH3 to both the Golgi and the cleavage furrow. Moreover telophase cells from mutants with defective GOLPH3-PI(4)P interaction fail to accumulate PI(4)P-and Rab11-associated secretory organelles at the cleavage site. Finally, we show that GOLPH3 protein interacts with components of both cytokinesis and membrane trafficking machineries in Drosophila cells. Based on these results we propose that GOLPH3 acts as a key molecule to coordinate phosphoinositide signaling with actomyosin dynamics and vesicle trafficking during cytokinesis. Because cytokinesis failures have been associated with premalignant disease and cancer, our studies suggest novel insight into molecular circuits involving the oncogene GOLPH3 in cytokinesis.

    View details for DOI 10.1371/journal.pgen.1004305

    View details for Web of Science ID 000337145100011

    View details for PubMedID 24786584

    View details for PubMedCentralID PMC4006750

  • The actin-binding protein profilin is required for germline stem cell maintenance and germ cell enclosure by somatic cyst cells DEVELOPMENT Shields, A. R., Spence, A. C., Yamashita, Y. M., Davies, E. L., Fuller, M. T. 2014; 141 (1): 73-82

    Abstract

    Specialized microenvironments, or niches, provide signaling cues that regulate stem cell behavior. In the Drosophila testis, the JAK-STAT signaling pathway regulates germline stem cell (GSC) attachment to the apical hub and somatic cyst stem cell (CySC) identity. Here, we demonstrate that chickadee, the Drosophila gene that encodes profilin, is required cell autonomously to maintain GSCs, possibly facilitating localization or maintenance of E-cadherin to the GSC-hub cell interface. Germline specific overexpression of Adenomatous Polyposis Coli 2 (APC2) rescued GSC loss in chic hypomorphs, suggesting an additive role of APC2 and F-actin in maintaining the adherens junctions that anchor GSCs to the niche. In addition, loss of chic function in the soma resulted in failure of somatic cyst cells to maintain germ cell enclosure and overproliferation of transit-amplifying spermatogonia.

    View details for DOI 10.1242/dev.101931

    View details for PubMedID 24346697

    View details for PubMedCentralID PMC3865751

  • The histone variant His2Av is required for adult stem cell maintenance in the Drosophila testis. PLoS genetics Morillo Prado, J. R., Srinivasan, S., Fuller, M. T. 2013; 9 (11)

    Abstract

    Many tissues are sustained by adult stem cells, which replace lost cells by differentiation and maintain their own population through self-renewal. The mechanisms through which adult stem cells maintain their identity are thus important for tissue homeostasis and repair throughout life. Here, we show that a histone variant, His2Av, is required cell autonomously for maintenance of germline and cyst stem cells in the Drosophila testis. The ATP-dependent chromatin-remodeling factor Domino is also required in this tissue for adult stem cell maintenance possibly by regulating the incorporation of His2Av into chromatin. Interestingly, although expression of His2Av was ubiquitous, its function was dispensable for germline and cyst cell differentiation, suggesting a specific role for this non-canonical histone in maintaining the stem cell state in these lineages.

    View details for DOI 10.1371/journal.pgen.1003903

    View details for PubMedID 24244183

    View details for PubMedCentralID PMC3820763

  • The Histone Variant His2Av is Required for Adult Stem Cell Maintenance in the Drosophila Testis. PLoS genetics Morillo Prado, J. R., Srinivasan, S., Fuller, M. T. 2013; 9 (11)

    View details for DOI 10.1371/journal.pgen.1003903

    View details for PubMedID 24244183

  • The polyubiquitin gene Ubi-p63E is essential for male meiotic cell cycle progression and germ cell differentiation in Drosophila. Development Lu, C., Kim, J., Fuller, M. T. 2013; 140 (17): 3522-3531

    Abstract

    The ubiquitin proteasome system (UPS) regulates many biological pathways by post-translationally ubiquitylating proteins for degradation. Although maintaining a dynamic balance between free ubiquitin and ubiquitylated proteins is key to UPS function, the mechanisms that regulate ubiquitin homeostasis in different tissues through development are not clear. Here we show, via analysis of the magellan (magn) complementation group, that loss of function of the Drosophila polyubiquitin Ubi-p63E results specifically in meiotic arrest sterility in males. Ubi-p63E contributes predominantly to maintaining the free ubiquitin pool in testes. The function of Ubi-p63E is required cell-autonomously for proper meiotic chromatin condensation, cell cycle progression and spermatid differentiation. magn mutant germ cells develop normally to the spermatocyte stage but arrest at the G2/M transition of meiosis I, with lack of protein expression of the key meiotic cell cycle regulators Boule and Cyclin B. Loss of Ubi-p63E function did not strongly affect the spermatocyte transcription program regulated by the testis TBP-associated factor (tTAF) or meiosis arrest complex (tMAC) genes. Knocking down proteasome function specifically in spermatocytes caused a different meiotic arrest phenotype, suggesting that the magn phenotype might not result from general defects in protein degradation. Our results suggest a conserved role of polyubiquitin genes in male meiosis and a potential mechanism leading to meiosis I maturation arrest.

    View details for DOI 10.1242/dev.098947

    View details for PubMedID 23884444

    View details for PubMedCentralID PMC3742140

  • Three levels of regulation lead to protamine and Mst77F expression in Drosophila DEVELOPMENTAL BIOLOGY Barckmann, B., Chen, X., Kaiser, S., Jayaramaiah-Raja, S., Rathke, C., Dottermusch-Heidel, C., Fuller, M. T., Renkawitz-Pohl, R. 2013; 377 (1): 33-45

    Abstract

    Differentiation from a haploid round spermatid to a highly streamlined, motile sperm requires temporal and spatial regulation of the expression of numerous proteins. One form of regulation is the storage of translationally repressed mRNAs. In Drosophila spermatocytes, the transcription of many of these translationally delayed mRNAs during spermiogenesis is in turn directly or indirectly regulated by testis-specific homologs of TATA-box-binding-protein-associated factors (tTAFs). Here we present evidence that expression of Mst77F, which is a specialized linker histone-like component of sperm chromatin, and of protamine B (ProtB), which contributes to formation of condensed sperm chromatin, is regulated at three levels. Transcription of Mst77F is guided by a short, promoter-proximal region, while expression of the Mst77F protein is regulated at two levels, early by translational repression via sequences mainly in the 5' part of the ORF and later by either protein stabilization or translational activation, dependent on sequences in the ORF. The protB gene is a direct target of tTAFs, with very short upstream regulatory regions of protB (-105 to +94 bp) sufficient for both cell-type-specific transcription and repression of translation in spermatocytes. In addition, efficient accumulation of the ProtB protein in late elongating spermatids depends on sequences in the ORF. We present evidence that spermatocytes provide the transacting mechanisms for translational repression of these mRNAs, while spermatids contain the machinery to activate or stabilize protamine accumulation for sperm chromatin components. Thus, the proper spatiotemporal expression pattern of major sperm chromatin components depends on cell-type-specific mechanisms of transcriptional and translational control.

    View details for DOI 10.1016/j.ydbio.2013.02.018

    View details for Web of Science ID 000318757300004

    View details for PubMedID 23466740

    View details for PubMedCentralID PMC4154633

  • The transcriptional regulator lola is required for stem cell maintenance and germ cell differentiation in the Drosophila testis DEVELOPMENTAL BIOLOGY Davies, E. L., Lim, J. G., Joo, W. J., Tam, C. H., Fuller, M. T. 2013; 373 (2): 310-321

    Abstract

    Stem cell behavior is regulated by extrinsic signals from specialized microenvironments, or niches, and intrinsic factors required for execution of context-appropriate responses to niche signals. Here we show that function of the transcriptional regulator longitudinals lacking (lola) is required cell autonomously for germline stem cell and somatic cyst stem cell maintenance in the Drosophila testis. In addition, lola is also required for proper execution of key developmental transitions during male germ cell differentiation, including the switch from transit amplifying progenitor to spermatocyte growth and differentiation, as well as meiotic cell cycle progression and spermiogenesis. Different lola isoforms, each having unique C-termini and zinc finger domains, may control different aspects of proliferation and differentiation in the male germline and somatic cyst stem cell lineages.

    View details for DOI 10.1016/j.ydbio.2012.11.004

    View details for Web of Science ID 000313381200008

    View details for PubMedID 23159836

  • Polycomb Group Genes Psc and Su(z)2 Maintain Somatic Stem Cell Identity and Activity in Drosophila PLOS ONE Prado, J. R., Chen, X., Fuller, M. T. 2012; 7 (12)

    Abstract

    Adult stem cells are essential for the proper function of many tissues, yet the mechanisms that maintain the proper identity and regulate proliferative capacity in stem cell lineages are not well understood. Polycomb group (PcG) proteins are transcriptional repressors that have recently emerged as important regulators of stem cell maintenance and differentiation. Here we describe the role of Polycomb Repressive Complex 1 (PRC1) genes Posterior sex combs (Psc) and Suppressor of zeste two (Su(z)2) in restricting the proliferation and maintaining the identity of the Cyst Stem Cell (CySC) lineage in the Drosophila testis. In contrast, Psc and Su(z)2 seem to be dispensable for both germline stem cell (GSC) maintenance and germ cell development. We show that loss of Psc and Su(z)2 function in the CySC lineage results in the formation of aggregates of mutant cells that proliferate abnormally, and display abnormal somatic identity correlated with derepression of the Hox gene Abdominal-B. Furthermore, we show that tumorigenesis in the CySC lineage interferes non-cell autonomously with maintenance of GSCs most likely by displacing them from their niche.

    View details for DOI 10.1371/journal.pone.0052892

    View details for Web of Science ID 000313158800106

    View details for PubMedCentralID PMC3528704

  • Mutations in Cog7 affect Golgi structure, meiotic cytokinesis and sperm development during Drosophila spermatogenesis JOURNAL OF CELL SCIENCE Belloni, G., Sechi, S., Riparbelli, M. G., Fuller, M. T., Callaini, G., Giansanti, M. G. 2012; 125 (22): 5441-5452

    Abstract

    The conserved oligomeric Golgi (COG) complex plays essential roles in Golgi function, vesicle trafficking and glycosylation. Deletions in the human COG7 gene are associated with a rare multisystemic congenital disorder of glycosylation that causes mortality within the first year of life. In this paper, we characterise the Drosophila orthologue of COG7 (Cog7). Loss-of-function Cog7 mutants are viable but male sterile. The Cog7 gene product is enriched in the Golgi stacks and in Golgi-derived structures throughout spermatogenesis. Mutations in the Cog7 gene disrupt Golgi architecture and reduce the number of Golgi stacks in primary spermatocytes. During spermiogenesis, loss of the Cog7 protein impairs the assembly of the Golgi-derived acroblast in spermatids and affects axoneme architecture. Similar to the Cog5 homologue, four way stop (Fws), Cog7 enables furrow ingression during cytokinesis. We show that the recruitment of the small GTPase Rab11 and the phosphatidylinositol transfer protein Giotto (Gio) to the cleavage site requires a functioning wild-type Cog7 gene. In addition, Gio coimmunoprecipitates with Cog7 and with Rab11 in the testes. Our results altogether implicate Cog7 as an upstream component in a gio-Rab11 pathway controlling membrane addition during cytokinesis.

    View details for DOI 10.1242/jcs.108878

    View details for Web of Science ID 000314511900018

    View details for PubMedID 22946051

  • What Drosophila Spermatocytes Tell Us About the Mechanisms Underlying Cytokinesis CYTOSKELETON Giansanti, M. G., Fuller, M. T. 2012; 69 (11): 869-881

    Abstract

    Cytokinesis separates the genomic material and organelles of a dividing cell equitably into two physically distinct daughter cells at the end of cell division. This highly choreographed process involves coordinated reorganization and regulated action of the actin and microtubule cytoskeletal systems, an assortment of motor proteins, and membrane trafficking components. Due to their large size, the ease with which exquisite cytological analysis may be performed on them, and the availability of numerous mutants and other genetic tools, Drosophila spermatocytes have provided an excellent system for exploring the mechanistic basis for the temporally programmed and precise spatially localized events of cytokinesis. Mutants defective in male meiotic cytokinesis can be easily identified in forward genetic screens by the production of multinucleate spermatids. In addition, the weak spindle assembly checkpoint in spermatocytes, which causes only a small delay of anaphase onset in the presence of unattached chromosomes, allows investigation of whether gene products required for spindle assembly and chromosome segregation are also involved in cytokinesis. Perhaps due to the large size of spermatocytes and the requirement for two rapid-fire rounds of division without intervening S or growth phases during meiosis, male meiotic mutants have also revealed much about molecular mechanisms underlying new membrane addition during cytokinesis. Finally, cell type-specific differences in the events that set up and complete cytokinesis are emerging from comparison of spermatocytes with cells undergoing mitosis either elsewhere in the organism or in tissue culture.

    View details for DOI 10.1002/cm.21063

    View details for Web of Science ID 000313747400002

    View details for PubMedID 22927345

  • A Novel Human Polycomb Binding Site Acts As a Functional Polycomb Response Element in Drosophila PLOS ONE Cuddapah, S., Roh, T., Cui, K., Jose, C. C., Fuller, M. T., Zhao, K., Chen, X. 2012; 7 (5)

    Abstract

    Polycomb group (PcG) proteins are key chromatin regulators implicated in multiple processes including embryonic development, tissue homeostasis, genomic imprinting, X-chromosome inactivation, and germ cell differentiation. The PcG proteins recognize target genomic loci through cis DNA sequences known as Polycomb Response Elements (PREs), which are well characterized in Drosophila. However, mammalian PREs have been elusive until two groups reported putative mammalian PREs recently. Consistent with the existence of mammalian PREs, here we report the identification and characterization of a potential PRE from human T cells. The putative human PRE has enriched binding of PcG proteins, and such binding is dependent on a key PcG component SUZ12. We demonstrate that the putative human PRE carries both genetic and molecular features of Drosophila PRE in transgenic flies, implying that not only the trans PcG proteins but also certain features of the cis PREs are conserved between mammals and Drosophila.

    View details for DOI 10.1371/journal.pone.0036365

    View details for Web of Science ID 000305343400023

    View details for PubMedID 22570707

    View details for PubMedCentralID PMC3343078

  • The receptor tyrosine phosphatase Lar regulates adhesion between Drosophila male germline stem cells and the niche DEVELOPMENT Srinivasan, S., Mahowald, A. P., Fuller, M. T. 2012; 139 (8): 1381-1390

    Abstract

    The stem cell niche provides a supportive microenvironment to maintain adult stem cells in their undifferentiated state. Adhesion between adult stem cells and niche cells or the local basement membrane ensures retention of stem cells in the niche environment. Drosophila male germline stem cells (GSCs) attach to somatic hub cells, a component of their niche, through E-cadherin-mediated adherens junctions, and orient their centrosomes toward these localized junctional complexes to carry out asymmetric divisions. Here we show that the transmembrane receptor tyrosine phosphatase Leukocyte-antigen-related-like (Lar), which is best known for its function in axonal migration and synapse morphogenesis in the nervous system, helps maintain GSCs at the hub by promoting E-cadherin-based adhesion between hub cells and GSCs. Lar is expressed in GSCs and early spermatogonial cells and localizes to the hub-GSC interface. Loss of Lar function resulted in a reduced number of GSCs at the hub. Lar function was required cell-autonomously in germ cells for proper localization of Adenomatous polyposis coli 2 and E-cadherin at the hub-GSC interface and for the proper orientation of centrosomes in GSCs. Ultrastructural analysis revealed that in Lar mutants the adherens junctions between hub cells and GSCs lack the characteristic dense staining seen in wild-type controls. Thus, the Lar receptor tyrosine phosphatase appears to polarize and retain GSCs through maintenance of localized E-cadherin-based adherens junctions.

    View details for DOI 10.1242/dev.070052

    View details for Web of Science ID 000301945100003

    View details for PubMedID 22378638

    View details for PubMedCentralID PMC3308176

  • Polycomb group genes Psc and Su(z)2 maintain somatic stem cell identity and activity in Drosophila. PloS one Morillo Prado, J. R., Chen, X., Fuller, M. T. 2012; 7 (12)

    Abstract

    Adult stem cells are essential for the proper function of many tissues, yet the mechanisms that maintain the proper identity and regulate proliferative capacity in stem cell lineages are not well understood. Polycomb group (PcG) proteins are transcriptional repressors that have recently emerged as important regulators of stem cell maintenance and differentiation. Here we describe the role of Polycomb Repressive Complex 1 (PRC1) genes Posterior sex combs (Psc) and Suppressor of zeste two (Su(z)2) in restricting the proliferation and maintaining the identity of the Cyst Stem Cell (CySC) lineage in the Drosophila testis. In contrast, Psc and Su(z)2 seem to be dispensable for both germline stem cell (GSC) maintenance and germ cell development. We show that loss of Psc and Su(z)2 function in the CySC lineage results in the formation of aggregates of mutant cells that proliferate abnormally, and display abnormal somatic identity correlated with derepression of the Hox gene Abdominal-B. Furthermore, we show that tumorigenesis in the CySC lineage interferes non-cell autonomously with maintenance of GSCs most likely by displacing them from their niche.

    View details for DOI 10.1371/journal.pone.0052892

    View details for PubMedID 23285219

    View details for PubMedCentralID PMC3528704

  • Germline Stem Cells COLD SPRING HARBOR PERSPECTIVES IN BIOLOGY Spradling, A., Fuller, M. T., Braun, R. E., Yoshida, S. 2011; 3 (11)

    Abstract

    Sperm and egg production requires a robust stem cell system that balances self-renewal with differentiation. Self-renewal at the expense of differentiation can cause tumorigenesis, whereas differentiation at the expense of self-renewal can cause germ cell depletion and infertility. In most organisms, and sometimes in both sexes, germline stem cells (GSCs) often reside in a defined anatomical niche. Factors within the niche regulate a balance between GSC self-renewal and differentiation. Asymmetric division of the germline stem cell to form daughter cells with alternative fates is common. The exception to both these tendencies is the mammalian testis where there does not appear to be an obvious anatomical niche and where GSC homeostasis is likely accomplished by a stochastic balance of self-renewal and differentiation and not by regulated asymmetric cell division. Despite these apparent differences, GSCs in all organisms share many common mechanisms, although not necessarily molecules, to guarantee survival of the germline.

    View details for DOI 10.1101/cshperspect.a002642

    View details for Web of Science ID 000296695500001

    View details for PubMedID 21791699

  • Role of Survivin in cytokinesis revealed by a separation-of-function allele MOLECULAR BIOLOGY OF THE CELL Szafer-Glusman, E., Fuller, M. T., Giansanti, M. G. 2011; 22 (20): 3779-3790
  • Sequential changes at differentiation gene promoters as they become active in a stem cell lineage DEVELOPMENT Chen, X., Lu, C., Prado, J. R., Eun, S. H., Fuller, M. T. 2011; 138 (12): 2441-2450

    Abstract

    Transcriptional silencing of terminal differentiation genes by the Polycomb group (PcG) machinery is emerging as a key feature of precursor cells in stem cell lineages. How, then, is this epigenetic silencing reversed for proper cellular differentiation? Here, we investigate how the developmental program reverses local PcG action to allow expression of terminal differentiation genes in the Drosophila male germline stem cell (GSC) lineage. We find that the silenced state, set up in precursor cells, is relieved through developmentally regulated sequential events at promoters once cells commit to spermatocyte differentiation. The programmed events include global downregulation of Polycomb repressive complex 2 (PRC2) components, recruitment of hypophosphorylated RNA polymerase II (Pol II) to promoters, as well as the expression and action of testis-specific homologs of TATA-binding protein-associated factors (tTAFs). In addition, action of the testis-specific meiotic arrest complex (tMAC), a tissue-specific version of the MIP/dREAM complex, is required both for recruitment of tTAFs to target differentiation genes and for proper cell type-specific localization of PRC1 components and tTAFs within the spermatocyte nucleolus. Together, the action of the tMAC and tTAF cell type-specific chromatin and transcription machinery leads to loss of Polycomb and release of stalled Pol II from the terminal differentiation gene promoters, allowing robust transcription.

    View details for DOI 10.1242/dev.056572

    View details for Web of Science ID 000290909400005

    View details for PubMedID 21610025

    View details for PubMedCentralID PMC3100706

  • E-Cadherin Is Required for Centrosome and Spindle Orientation in Drosophila Male Germline Stem Cells PLOS ONE Inaba, M., Yuan, H., Salzmann, V., Fuller, M. T., Yamashita, Y. M. 2010; 5 (8)

    Abstract

    Many adult stem cells reside in a special microenvironment known as the niche, where they receive essential signals that specify stem cell identity. Cell-cell adhesion mediated by cadherin and integrin plays a crucial role in maintaining stem cells within the niche. In Drosophila melanogaster, male germline stem cells (GSCs) are attached to niche component cells (i.e., the hub) via adherens junctions. The GSC centrosomes and spindle are oriented toward the hub-GSC junction, where E-cadherin-based adherens junctions are highly concentrated. For this reason, adherens junctions are thought to provide a polarity cue for GSCs to enable proper orientation of centrosomes and spindles, a critical step toward asymmetric stem cell division. However, understanding the role of E-cadherin in GSC polarity has been challenging, since GSCs carrying E-cadherin mutations are not maintained in the niche. Here, we tested whether E-cadherin is required for GSC polarity by expressing a dominant-negative form of E-cadherin. We found that E-cadherin is indeed required for polarizing GSCs toward the hub cells, an effect that may be mediated by Apc2. We also demonstrated that E-cadherin is required for the GSC centrosome orientation checkpoint, which prevents mitosis when centrosomes are not correctly oriented. We propose that E-cadherin orchestrates multiple aspects of stem cell behavior, including polarization of stem cells toward the stem cell-niche interface and adhesion of stem cells to the niche supporting cells.

    View details for DOI 10.1371/journal.pone.0012473

    View details for Web of Science ID 000281405300011

    View details for PubMedID 20824213

  • The Drosophila SUN protein Spag4 cooperates with the coiled-coil protein Yuri Gagarin to maintain association of the basal body and spermatid nucleus JOURNAL OF CELL SCIENCE Kracklauer, M. P., Wiora, H. M., Deery, W. J., Chen, X., Bolival, B., Romanowicz, D., Simonette, R. A., Fuller, M. T., Fischer, J. A., Beckingham, K. M. 2010; 123 (16): 2763-2772

    Abstract

    Maintaining the proximity of centrosomes to nuclei is important in several cellular contexts, and LINC complexes formed by SUN and KASH proteins are crucial in this process. Here, we characterize the presumed Drosophila ortholog of the mammalian SUN protein, sperm-associated antigen 4 (Spag4, previously named Giacomo), and demonstrate that Spag4 is required for centriole and nuclear attachment during spermatogenesis. Production of spag4 mRNA is limited to the testis, and Spag4 protein shows a dynamic pattern of association with the germline nuclei, including a concentration of protein at the site of attachment of the single spermatid centriole. In the absence of Spag4, nuclei and centrioles or basal bodies (BBs) dissociate from each other after meiosis. This role of Spag4 in centriolar attachment does not involve either of the two KASH proteins of the Drosophila genome (Klarsicht and MSP-300), but does require the coiled-coil protein Yuri Gagarin. Yuri shows an identical pattern of localization at the nuclear surface to Spag4 during spermatogenesis, and epistasis studies show that the activities of Yuri and dynein-dynactin are downstream of spag4 in this centriole attachment pathway. The later defects in spermatogenesis seen for yuri and spag4 mutants are similar, suggesting they could be secondary to initial disruption of events at the nuclear surface.

    View details for DOI 10.1242/jcs.066589

    View details for Web of Science ID 000280622700009

    View details for PubMedID 20647369

    View details for PubMedCentralID PMC2915878

  • Phosphatidylinositol 4,5-bisphosphate Directs Spermatid Cell Polarity and Exocyst Localization in Drosophila MOLECULAR BIOLOGY OF THE CELL Fabian, L., Wei, H., Rollins, J., Noguchi, T., Blankenship, J. T., Bellamkonda, K., Polevoy, G., Gervais, L., Guichet, A., Fuller, M. T., Brill, J. A. 2010; 21 (9): 1546-1555

    Abstract

    During spermiogenesis, Drosophila melanogaster spermatids coordinate their elongation in interconnected cysts that become highly polarized, with nuclei localizing to one end and sperm tail growth occurring at the other. Remarkably little is known about the signals that drive spermatid polarity and elongation. Here we identify phosphoinositides as critical regulators of these processes. Reduction of plasma membrane phosphatidylinositol 4,5-bisphosphate (PIP(2)) by low-level expression of the PIP(2) phosphatase SigD or mutation of the PIP(2) biosynthetic enzyme Skittles (Sktl) results in dramatic defects in spermatid cysts, which become bipolar and fail to fully elongate. Defects in polarity are evident from the earliest stages of elongation, indicating that phosphoinositides are required for establishment of polarity. Sktl and PIP(2) localize to the growing end of the cysts together with the exocyst complex. Strikingly, the exocyst becomes completely delocalized when PIP(2) levels are reduced, and overexpression of Sktl restores exocyst localization and spermatid cyst polarity. Moreover, the exocyst is required for polarity, as partial loss of function of the exocyst subunit Sec8 results in bipolar cysts. Our data are consistent with a mechanism in which localized synthesis of PIP(2) recruits the exocyst to promote targeted membrane delivery and polarization of the elongating cysts.

    View details for DOI 10.1091/mbc.E09-07-0582

    View details for Web of Science ID 000277179600011

    View details for PubMedID 20237161

  • Accumulation of a differentiation regulator specifies transit amplifying division number in an adult stem cell lineage PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Insco, M. L., Leon, A., Tam, C. H., McKearin, D. M., Fuller, M. T. 2009; 106 (52): 22311-22316

    Abstract

    A key feature of many adult stem cell lineages is that stem cell daughters destined for differentiation undergo several transit amplifying (TA) divisions before initiating terminal differentiation, allowing few and infrequently dividing stem cells to produce many differentiated progeny. Although the number of progenitor divisions profoundly affects tissue (re)generation, and failure to control these divisions may contribute to cancer, the mechanisms that limit TA proliferation are not well understood. Here, we use a model stem cell lineage, the Drosophila male germ line, to investigate the mechanism that counts the number of TA divisions. The Drosophila Bag of Marbles (Bam) protein is required for male germ cells to cease spermatogonial TA divisions and initiate spermatocyte differentiation [McKearin DM, et al. (1990) Genes Dev 4:2242-2251]. Contrary to models involving dilution of a differentiation repressor, our results suggest that the switch from proliferation to terminal differentiation is triggered by accumulation of Bam protein to a critical threshold in TA cells and that the number of TA divisions is set by the timing of Bam accumulation with respect to the rate of cell cycle progression.

    View details for DOI 10.1073/pnas.0912454106

    View details for Web of Science ID 000273178700050

    View details for PubMedID 20018708

    View details for PubMedCentralID PMC2799733

  • TRAPPII is required for cleavage furrow ingression and localization of Rab11 in dividing male meiotic cells of Drosophila JOURNAL OF CELL SCIENCE Robinett, C. C., Giansanti, M. G., Gatti, M., Fuller, M. T. 2009; 122 (24): 4526-4534

    Abstract

    Although membrane addition is crucial for cytokinesis in many animal cell types, the specific mechanisms supporting cleavage furrow ingression are not yet understood. Mutations in the gene brunelleschi (bru), which encodes the Drosophila ortholog of the yeast Trs120p subunit of TRAPPII, cause failure of furrow ingression in male meiotic cells. In non-dividing cells, Brunelleschi protein fused to GFP is dispersed throughout the cytoplasm and enriched at Golgi organelles, similarly to another Drosophila TRAPPII subunit, dBet3. Localization of the membrane-trafficking GTPase Rab11 to the cleavage furrow requires wild-type function of bru, and genetic interactions between bru and Rab11 increase the failure of meiotic cytokinesis and cause synthetic lethality. bru also genetically interacts with four wheel drive (fwd), which encodes a PI4Kbeta, such that double mutants exhibit enhanced failure of male meiotic cytokinesis. These results suggest that Bru cooperates with Rab11 and PI4Kbeta to regulate the efficiency of membrane addition to the cleavage furrow, thus promoting cytokinesis in Drosophila male meiotic cells.

    View details for DOI 10.1242/jcs.054536

    View details for Web of Science ID 000272329300013

    View details for PubMedID 19934220

    View details for PubMedCentralID PMC2787463

  • Regulation of self-renewal, proliferation and differentiation in an adult stem cell lineage 16th Annual Conference of the International-Society-of-Development-Biologists Fuller, M. T., Davies, E., Spence, A. ELSEVIER SCIENCE BV. 2009: S3–S3
  • Molecular Evolution of the Testis TAFs of Drosophila MOLECULAR BIOLOGY AND EVOLUTION Li, V. C., Davis, J. C., Lenkov, K., Bolival, B., Fuller, M. T., Petrov, D. A. 2009; 26 (5): 1103-1116

    Abstract

    The basal transcription machinery is responsible for initiating transcription at core promoters. During metazoan evolution, its components have expanded in number and diversified to increase the complexity of transcriptional regulation in tissues and developmental stages. To explore the evolutionary events and forces underlying this diversification, we analyzed the evolution of the Drosophila testis TAFs (TBP-associated factors), paralogs of TAFs from the basal transcription factor TFIID that are essential for normal transcription during spermatogenesis of a large set of specific genes involved in terminal differentiation of male gametes. There are five testis-specific TAFs in Drosophila, each expressed only in primary spermatocytes and each a paralog of a different generally expressed TFIID subunit. An examination of the presence of paralogs across taxa as well as molecular clock dating indicates that all five testis TAFs likely arose within a span of approximately 38 My 63-250 Ma by independent duplication events from their generally expressed paralogs. Furthermore, the evolution of the testis TAFs has been rapid, with apparent further accelerations in multiple Drosophila lineages. Analysis of between-species divergence and intraspecies polymorphism indicates that the major forces of evolution on these genes have been reduced purifying selection, pervasive positive selection, and coevolution. Other genes that exhibit similar patterns of evolution in the Drosophila lineages are also characterized by enriched expression in the testis, suggesting that the pervasive positive selection acting on the tTAFs is likely to be related to their expression in the testis.

    View details for DOI 10.1093/molbev/msp030

    View details for Web of Science ID 000265274000014

    View details for PubMedID 19244474

    View details for PubMedCentralID PMC2727373

  • Centrosome misorientation reduces stem cell division during ageing NATURE Cheng, J., Turkel, N., Hemati, N., Fuller, M. T., Hunt, A. J., Yamashita, Y. M. 2008; 456 (7222): 599-U40

    Abstract

    Asymmetric division of adult stem cells generates one self-renewing stem cell and one differentiating cell, thereby maintaining tissue homeostasis. A decline in stem cell function has been proposed to contribute to tissue ageing, although the underlying mechanism is poorly understood. Here we show that changes in the stem cell orientation with respect to the niche during ageing contribute to the decline in spermatogenesis in the male germ line of Drosophila. Throughout the cell cycle, centrosomes in germline stem cells (GSCs) are oriented within their niche and this ensures asymmetric division. We found that GSCs containing misoriented centrosomes accumulate with age and that these GSCs are arrested or delayed in the cell cycle. The cell cycle arrest is transient, and GSCs appear to re-enter the cell cycle on correction of centrosome orientation. On the basis of these findings, we propose that cell cycle arrest associated with centrosome misorientation functions as a mechanism to ensure asymmetric stem cell division, and that the inability of stem cells to maintain correct orientation during ageing contributes to the decline in spermatogenesis. We also show that some of the misoriented GSCs probably originate from dedifferentiation of spermatogonia.

    View details for DOI 10.1038/nature07386

    View details for Web of Science ID 000261340000032

    View details for PubMedID 18923395

  • A role for very-long-chain fatty acids in furrow ingression during cytokinesis in Drosophila spermatocytes CURRENT BIOLOGY Szafer-Glusman, E., Giansanti, M. G., Nishihama, R., Bolival, B., Pringle, J., Gatti, M., Fuller, M. T. 2008; 18 (18): 1426-1431

    Abstract

    Cell shape and membrane remodeling rely on regulated interactions between the lipid bilayer and cytoskeletal arrays at the cell cortex. During cytokinesis, animal cells build an actomyosin ring anchored to the plasma membrane at the equatorial cortex. Ring constriction coupled to plasma-membrane ingression separates the two daughter cells. Plasma-membrane lipids influence membrane biophysical properties such as membrane curvature and elasticity and play an active role in cell function, and specialized membrane domains are emerging as important factors in regulating assembly and rearrangement of the cytoskeleton. Here, we show that mutations in the gene bond, which encodes a Drosophila member of the family of Elovl proteins that mediate elongation of very-long-chain fatty acids, block or dramatically slow cleavage-furrow ingression during early telophase in dividing spermatocytes. In bond mutant cells at late stages of division, the contractile ring frequently detaches from the cortex and constricts or collapses to one side of the cell, and the cleavage furrow regresses. Our findings implicate very-long-chain fatty acids or their derivative complex lipids in allowing supple membrane deformation and the stable connection of cortical contractile components to the plasma membrane during cell division.

    View details for DOI 10.1016/j.cub.2008.08.061

    View details for Web of Science ID 000259523600030

    View details for PubMedID 18804373

    View details for PubMedCentralID PMC2577570

  • Moesin and its activating kinase Slik are required for cortical stability and microtubule organization in mitotic cells JOURNAL OF CELL BIOLOGY Carreno, S., Kouranti, I., Glusman, E. S., Fuller, M. T., Echard, A., Payre, F. 2008; 180 (4): 739-746

    Abstract

    Cell division requires cell shape changes involving the localized reorganization of cortical actin, which must be tightly linked with chromosome segregation operated by the mitotic spindle. How this multistep process is coordinated remains poorly understood. In this study, we show that the actin/membrane linker moesin, the single ERM (ezrin, radixin, and moesin) protein in Drosophila melanogaster, is required to maintain cortical stability during mitosis. Mitosis onset is characterized by a burst of moesin activation mediated by a Slik kinase-dependent phosphorylation. Activated moesin homogenously localizes at the cortex in prometaphase and is progressively restricted at the equator in later stages. Lack of moesin or inhibition of its activation destabilized the cortex throughout mitosis, resulting in severe cortical deformations and abnormal distribution of actomyosin regulators. Inhibiting moesin activation also impaired microtubule organization and precluded stable positioning of the mitotic spindle. We propose that the spatiotemporal control of moesin activation at the mitotic cortex provides localized cues to coordinate cortical contractility and microtubule interactions during cell division.

    View details for DOI 10.1083/jcb.200709161

    View details for Web of Science ID 000253494000024

    View details for PubMedID 18283112

  • Asymmetric centrosome behavior and the mechanisms of stem cell division JOURNAL OF CELL BIOLOGY Yamashita, Y. M., Fuller, M. T. 2008; 180 (2): 261-266

    Abstract

    The ability of dividing cells to produce daughters with different fates is an important developmental mechanism conserved from bacteria to fungi, plants, and metazoan animals. Asymmetric outcomes of a cell division can be specified by two general mechanisms: asymmetric segregation of intrinsic fate determinants or asymmetric placement of daughter cells into microenvironments that provide extrinsic signals that direct cells to different states. For both, spindle orientation must be coordinated with the localization of intrinsic determinants or source of extrinsic signals to achieve the proper asymmetric outcome. Recent work on spindle orientation in Drosophila melanogaster male germline stem cells and neuroblasts has brought into sharp focus the key role of differential centrosome behavior in developmentally programmed asymmetric division (for reviews see Cabernard, C., and C.Q. Doe. 2007. Curr. Biol. 17:R465-R467; Gonzalez, C. 2007. Nat. Rev. Genet. 8:462-472). These findings provide new insights and suggest intriguing new models for how cells coordinate spindle orientation with their cellular microenvironment to regulate and direct cell fate decisions within tissues.

    View details for Web of Science ID 000252746900004

    View details for PubMedID 18209101

  • Regulation of Self-renewal and Differentiation in Adult Stem Cell Lineages: Lessons from the Drosophila Male Germ Line 73rd Cold Spring Harbor Symposium on Quantitative Biology Davies, E. L., Fuller, M. T. COLD SPRING HARBOR LABORATORY PRESS. 2008: 137–145

    Abstract

    The ability to identify stem cells and trace their descendants in vivo has yielded insights into how self-renewal, proliferation, and differentiation are regulated in adult stem cell lineages. Analysis of male germ-line stem cells in Drosophila has revealed the importance of local signals from the microenvironment, the stem cell niche, in controlling stem cell behavior. Germ-line stem cells physically attach to the niche via localized adherens junctions that provide a polarity cue for orientation of centrosomes in interphase and the spindle in mitosis. As a result, stem cells divide asymmetrically: One daughter inherits attachment to the niche and remains within its embrace, whereas the other is displaced away and initiates differentiation. Strikingly, much as leukemia inhibitory factor (LIF) and transforming growth factor-beta (TGF-beta) signaling maintain mouse embryonic stem (ES) cells, maintenance of stem cell state in the Drosophila male germ line is regulated by cytokine-like signals from hub cells that activate the transcription factor STAT (signal transducer and activator of transcription) and TGF-beta class signals from surrounding support cells that repress expression of a key differentiation factor. Surprisingly, transit-amplifying cells can revert to the stem cell state if they reoccupy the niche. Upon cessation of mitosis and the switch to terminal differentiation, germ cells express cell-type- and stage-specific transcription machinery components that drive expression of terminal differentiation genes, in part by removing Polycomb transcriptional silencing machinery.

    View details for Web of Science ID 000267135700017

    View details for PubMedID 19329574

  • Centrosome misorientation reduces stem cell division during ageing. Nature Cheng, J., N. Turkel, N. Hemati, M. T. Fuller, A. J. Hunt, Y. M. Yamashita 2008; 456: 599-604
  • The Drosophila homolog of the Exo84 exocyst subunit promotes apical epithelial identity JOURNAL OF CELL SCIENCE Blankenship, J. T., Fuller, M. T., Zallen, J. A. 2007; 120 (17): 3099-3110

    Abstract

    The polarized architecture of epithelial tissues involves a dynamic balance between apical and basolateral membrane domains. Here we show that epithelial polarity in the Drosophila embryo requires the exocyst complex subunit homolog Exo84. Exo84 activity is essential for the apical localization of the Crumbs transmembrane protein, a key determinant of epithelial apical identity. Adherens junction proteins become mislocalized at the cell surface in Exo84 mutants in a pattern characteristic of defects in apical, but not basolateral, components. Loss of Crumbs from the cell surface precedes the disruption of Bazooka and Armadillo localization in Exo84 mutants. Moreover, Exo84 mutants display defects in apical cuticle secretion that are similar to crumbs mutants and are suppressed by a reduction in the basolateral proteins Dlg and Lgl. In Exo84 mutants at advanced stages of epithelial degeneration, apical and adherens junction proteins accumulate in an expanded recycling endosome compartment. These results suggest that epithelial polarity in the Drosophila embryo is actively maintained by exocyst-dependent apical localization of the Crumbs transmembrane protein.

    View details for DOI 10.1242/jcs.004770

    View details for Web of Science ID 000249559000012

    View details for PubMedID 17698923

  • Translational control of meiotic cell cycle progression and spermatid differentiation in male germ cells by a novel eIF4G homolog DEVELOPMENT Baker, C. C., Fuller, M. T. 2007; 134 (15): 2863-2869

    Abstract

    Translational control is crucial for proper timing of developmental events that take place in the absence of transcription, as in meiotic activation in oocytes, early embryogenesis in many organisms, and spermatogenesis. Here we show that a novel form of the translation initiation complex component eIF4G in Drosophila, eIF4G2, is required specifically for male germ cells to undergo meiotic division and proper spermatid differentiation. Flies mutant for eIF4G2 are viable and female fertile but male sterile. Spermatocytes form, but the germ cells in mutant males skip the major events of the meiotic divisions and form aberrant spermatids with large nuclei. Consistent with the failure to undergo the meiotic divisions, function of eIF4G2 is required post-transcriptionally for normal accumulation of the core cell cycle regulatory proteins Twine and CycB in mature spermatocytes. Loss of eIF4G2 function also causes widespread defects in spermatid differentiation. Although differentiation markers Dj and Fzo are expressed in late-stage eIF4G2 mutant germ cells, several key steps of spermatid differentiation fail, including formation of a compact mitochondrial derivative and full elongation. Our results suggest that an alternate form of the translation initiation machinery may be required for regulation and execution of key steps in male germ cell differentiation.

    View details for DOI 10.1242/dev.003764

    View details for Web of Science ID 000248381600015

    View details for PubMedID 17611220

  • Antagonistic roles of Rac and Rho in organizing the germ cell microenvironment CURRENT BIOLOGY Sarkar, A., Parikh, N., Hearn, S. A., Fuller, M. T., Tazuke, S. I., Schulz, C. 2007; 17 (14): 1253-1258

    Abstract

    The capacity of stem cells to self renew and the ability of stem cell daughters to differentiate into highly specialized cells depend on external cues provided by their cellular microenvironments [1-3]. However, how microenvironments are shaped is poorly understood. In testes of Drosophila melanogaster, germ cells are enclosed by somatic support cells. This physical interrelationship depends on signaling from germ cells to the Epidermal growth factor receptor (Egfr) on somatic support cells [4]. We show that germ cells signal via the Egf class ligand Spitz (Spi) and provide evidence that the Egfr associates with and acts through the guanine nucleotide exchange factor Vav to regulate activities of Rac1. Reducing activity of the Egfr, Vav, or Rac1 from somatic support cells enhanced the germ cell enclosure defects of a conditional spi allele. Conversely, reducing activity of Rho1 from somatic support cells suppressed the germ cell enclosure defects of the conditional spi allele. We propose that a differential in Rac and Rho activities across somatic support cells guides their growth around the germ cells. Our novel findings reveal how signals from one cell type regulate cell-shape changes in another to establish a critical partnership required for proper differentiation of a stem cell lineage.

    View details for DOI 10.1016/j.cub.2007.06.048

    View details for Web of Science ID 000248202600031

    View details for PubMedID 17629483

  • Male and female Drosophila germline stem cells: Two versions of immortality SCIENCE Fuller, M. T., Spradling, A. C. 2007; 316 (5823): 402-404

    Abstract

    Drosophila male and female germline stem cells (GSCs) are sustained by niches and regulatory pathways whose common principles serve as models for understanding mammalian stem cells. Despite striking cellular and genetic similarities that suggest a common evolutionary origin, however, male and female GSCs also display important differences. Comparing these two stem cells and their niches in detail is likely to reveal how a common heritage has been adapted to the differing requirements of male and female gamete production.

    View details for DOI 10.1126/science.1140861

    View details for Web of Science ID 000245813400041

    View details for PubMedID 17446390

  • Phosphorylation of histone H4 Ser1 regulates sporulation in yeast and is conserved in fly and mouse spermatogenesis GENES & DEVELOPMENT Krishnamoorthy, T., Chen, X., Govin, J., Cheung, W. L., Dorsey, J., Schindler, K., Winter, E., Allis, C. D., Guacci, V., Khochbin, S., Fuller, M. T., Berger, S. L. 2006; 20 (18): 2580-2592

    Abstract

    Sporulation in Saccharomyces cerevisiae is a highly regulated process wherein a diploid cell gives rise to four haploid gametes. In this study we show that histone H4 Ser1 is phosphorylated (H4 S1ph) during sporulation, starting from mid-sporulation and persisting to germination, and is temporally distinct from earlier meiosis-linked H3 S10ph involved in chromosome condensation. A histone H4 S1A substitution mutant forms aberrant spores and has reduced sporulation efficiency. Deletion of sporulation-specific yeast Sps1, a member of the Ste20 family of kinases, nearly abolishes the sporulation-associated H4 S1ph modification. H4 S1ph may promote chromatin compaction, since deletion of SPS1 increases accessibility to antibody immunoprecipitation; furthermore, either deletion of Sps1 or an H4 S1A substitution results in increased DNA volume in nuclei within spores. We find H4 S1ph present during Drosophila melanogaster and mouse spermatogenesis, and similar to yeast, this modification extends late into sperm differentiation relative to H3 S10ph. Thus, H4 S1ph may be an evolutionarily ancient histone modification to mark the genome for gamete-associated packaging.

    View details for DOI 10.1101/gad.1457006

    View details for Web of Science ID 000240526700010

    View details for PubMedID 16980586

    View details for PubMedCentralID PMC1578680

  • Stem cells and cancer: Two faces of eve CELL Clarke, M. F., Fuller, M. 2006; 124 (6): 1111-1115

    Abstract

    Recent evidence suggests that a subset of cancer cells within some tumors, the so-called cancer stem cells, may drive the growth and metastasis of these tumors. Understanding the pathways that regulate proliferation, self-renewal, survival, and differentiation of malignant and normal stem cells may shed light on mechanisms that lead to cancer and suggest better modes of treatment.

    View details for DOI 10.1016/j.cell.2006.03.011

    View details for Web of Science ID 000237241400007

    View details for PubMedID 16564000

  • Histone modifications in spermatogenesis 31st Annual Meeting of the American-Society-of-Andrology Chang, S. L., Chen, X., McCallum, S., Fuller, M. T. AMER SOC ANDROLOGY, INC. 2006: 34–34
  • The class IPITP giotto is required for Drosophila cytokinesis CURRENT BIOLOGY Giansanti, M. G., Bonaccorsi, S., Kurek, R., Farkas, R. M., Dimitri, P., Fuller, M. T., Gatti, M. 2006; 16 (2): 195-201

    Abstract

    Phosphatidylinositol transfer proteins (PITPs) are highly conserved polypeptides that bind phosphatidylinositol or phosphatidylcholine monomers, facilitating their transfer from one membrane compartment to another . Although PITPs have been implicated in a variety of cellular functions, including lipid-mediated signaling and membrane trafficking, the precise biological roles of most PITPs remain to be elucidated . Here we show for the first time that a class I PITP is involved in cytokinesis. We found that giotto (gio), a Drosophila gene that encodes a class I PITP, serves an essential function required for both mitotic and meiotic cytokinesis. Neuroblasts and spermatocytes from gio mutants both assemble regular actomyosin rings. However, these rings fail to constrict to completion, leading to cytokinesis failures. Moreover, gio mutations cause an abnormal accumulation of Golgi-derived vesicles at the equator of spermatocyte telophases, suggesting that Gio is implicated in membrane-vesicle fusion. Consistent with these results, we found that Gio is enriched at the cleavage furrow, the ER, and the spindle envelope. We propose that Gio mediates transfer of lipid monomers from the ER to the equatorial membrane, causing a specific local enrichment in phosphatidylinositol. This change in membrane composition would ultimately facilitate vesicle fusion, allowing membrane addition to the furrow and/or targeted delivery of proteins required for cytokinesis.

    View details for DOI 10.1016/j.cub.2005.12.011

    View details for Web of Science ID 000235105900025

    View details for PubMedID 16431372

  • Asymmetric stem cell division and function of the niche in the Drosophila male germ line INTERNATIONAL JOURNAL OF HEMATOLOGY Yamashita, Y. M., Fuller, M. T. 2005; 82 (5): 377-380

    Abstract

    The balance between stem cell and differentiating cell populations is critical for the long-term maintenance of tissue renewal for cell types derived from adult stem cell lineages such as blood, skin, intestinal epithelium, and sperm. To keep this balance, stem cells have the potential to divide asymmetrically, producing one daughter cell that maintains stem cell identity and one daughter cell that initiates differentiation. In many adult stem cell systems, the maintenance, proliferation, and number of stem cells appear to be controlled by the microenvironment, or niche. The Drosophila male and female germ line provide excellent model systems in which to study asymmetric stem cell divisions within the stem cell niche. In addition to signals from the niche that specify stem cell self-renewal, the stem cells themselves have elaborate cellular mechanisms to ensure the asymmetric outcome of cell division.

    View details for DOI 10.1532/IJH97.05097

    View details for Web of Science ID 000234398000002

    View details for PubMedID 16533738

  • Signaling in stem cell niches: lessons from the Drosophila germline JOURNAL OF CELL SCIENCE Yamashita, Y. M., Fuller, M. T., Jones, D. L. 2005; 118 (4): 665-672

    Abstract

    Stem cells are cells that, upon division, can produce new stem cells as well as daughter cells that initiate differentiation along a specific lineage. Studies using the Drosophila germline as a model system have demonstrated that signaling from the stem cell niche plays a crucial role in controlling stem cell behavior. Surrounding support cells secrete growth factors that activate signaling within adjacent stem cells to specify stem cell self-renewal and block differentiation. In addition, cell-cell adhesion between stem cells and surrounding support cells is important for holding stem cells close to self-renewal signals. Furthermore, a combination of localized signaling and autonomously acting proteins might polarize stem cells in such a way as to ensure asymmetric stem cell divisions. Recent results describing stem cell niches in other adult stem cells, including hematopoietic and neural stem cells, have demonstrated that the features characteristic of stem cell niches in Drosophila gonads might be conserved.

    View details for DOI 10.1242/jcs.01680

    View details for Web of Science ID 000227741500004

    View details for PubMedID 15701923

  • Belle is a Drosophila DEAD-box protein required for viability and in the germ line DEVELOPMENTAL BIOLOGY Johnstone, O., Deuring, R., Bock, R., Linder, P., Fuller, M. T., Lasko, P. 2005; 277 (1): 92-101

    Abstract

    DEAD-box proteins are ATP-dependent RNA helicases that function in various stages of RNA processing and in RNP remodeling. Here, we report identification and characterization of the Drosophila protein Belle (Bel), which belongs to a highly conserved subfamily of DEAD-box proteins including yeast Ded1p, Xenopus An3, mouse PL10, human DDX3/DBX, and human DBY. Mutations in DBY are a frequent cause of male infertility in humans. Bel can substitute in vivo for Ded1p, an essential yeast translation factor, suggesting a requirement for Bel in translation initiation. Consistent with an essential cellular function, strong loss of function mutations in bel are recessive lethal with a larval growth defect phenotype. Hypomorphic bel mutants are male-sterile. Bel is also closely related to the Drosophila DEAD-box protein Vasa (Vas), a germ line-specific translational regulator. We find that Bel and Vas colocalize in nuage and at the oocyte posterior during oogenesis, and that bel function is required for female fertility. However, unlike Vas, Bel is not specifically enriched in embryonic pole cells. We conclude that the DEAD-box protein Bel has evolutionarily conserved roles in fertility and development.

    View details for DOI 10.1016/j.ydbio.2004.09.009

    View details for Web of Science ID 000225741200008

    View details for PubMedID 15572142

  • Testis-specific TAF homologs collaborate to control a tissue-specific transcription program DEVELOPMENT Hiller, M., Chen, X., Pringle, M. J., Suchorolski, M., Sancak, Y., Viswanathan, S., Bolival, B., Lin, T. Y., Marino, S., Fuller, M. T. 2004; 131 (21): 5297-5308

    Abstract

    Alternate forms of the PolII transcription initiation machinery have been proposed to play a role in selective activation of cell-type-specific gene expression programs during cellular differentiation. The cannonball (can) gene of Drosophila encodes a homolog of a TBP-associated factor (dTAF5) protein expressed only in spermatocytes, where it is required for normal transcription of genes required for spermatid differentiation. We show that Drosophila primary spermatocytes also express four additional tissue-specific TAFs: nht (homolog of dTAF4), mia (homolog of dTAF6), sa (homolog of dTAF8) and rye (homolog of dTAF12). Mutations in nht, mia and sa have similar effects in primary spermatocytes on transcription of several target genes involved in spermatid differentiation, and cause the same phenotypes as mutations in can, blocking both meiotic cell cycle progression and spermatid differentiation. The nht, mia, sa and rye proteins contain histone fold domain dimerization motifs. The nht and rye proteins interact structurally when co-expressed in bacteria, similarly to their generally expressed homologs TAF4 and TAF12, which heterodimerize. Strikingly, the structural interaction is tissue specific: nht did not interact with dTAF12 and dTAF4 did not interact with rye in a bacterial co-expression assay. We propose that the products of the five Drosophila genes encoding testis TAF homologs collaborate in an alternative TAF-containing protein complex to regulate a testis-specific gene expression program in primary spermatocytes required for terminal differentiation of male germ cells.

    View details for Web of Science ID 000225422200009

    View details for PubMedID 15456720

  • Germ-line specific variants of components of the mitochondrial outer membrane import machinery in Drosophila FEBS LETTERS Hwa, J. J., Zhu, A. J., Hiller, M. A., Kon, C. Y., Fuller, M. T., Santel, A. 2004; 572 (1-3): 141-146

    Abstract

    A search of the Drosophila genome for genes encoding components of the mitochondrial translocase of outer membrane (TOM) complex revealed duplication of genes encoding homologues of Tom20 and Tom40. Tom20 and Tom40 were represented by two differentially expressed homologues in the Drosophila genome. While dtom20 and dtom40 appeared to be expressed ubiquitously, the second variants, called tomboy20 and tomboy40, were expressed only in the male germ-line. Transcripts for tomboy20 and tomboy40 were detected in primary spermatocytes as well as post-meiotic stages. Transcription of tomboy20 and tomboy40 in spermatocytes was not dependent on the transcription factor Cannonball, which is responsible for controlling expression of gene products exclusively required for post-meiotic germ cell differentiation. Epitope-tagging and transient expression of dTom20 and Tomboy40 in mammalian cell culture showed proper targeting to mitochondria.

    View details for DOI 10.1016/j.febslet.2004.07.025

    View details for Web of Science ID 000223519300026

    View details for PubMedID 15304338

  • A misexpression screen reveals effects of bag-of-marbles and TGF beta class signaling on the Drosophila male germ-line stem cell lineage GENETICS Schulz, C., Kiger, A. A., Tazuke, S. I., Yamashita, Y. M., Pantalena, L. C., Jones, D. L., Wood, C. G., Fuller, M. T. 2004; 167 (2): 707-723

    Abstract

    Male gametes are produced throughout reproductive life by a classic stem cell mechanism. However, little is known about the molecular mechanisms for lineage production that maintain male germ-line stem cell (GSC) populations, regulate mitotic amplification divisions, and ensure germ cell differentiation. Here we utilize the Drosophila system to identify genes that cause defects in the male GSC lineage when forcibly expressed. We conducted a gain-of-function screen using a collection of 2050 EP lines and found 55 EP lines that caused defects at early stages of spermatogenesis upon forced expression either in germ cells or in surrounding somatic support cells. Most strikingly, our analysis of forced expression indicated that repression of bag-of-marbles (bam) expression in male GSC is important for male GSC survival, while activity of the TGF beta signal transduction pathway may play a permissive role in maintenance of GSCs in Drosophila testes. In addition, forced activation of the TGF beta signal transduction pathway in germ cells inhibits the transition from the spermatogonial mitotic amplification program to spermatocyte differentiation.

    View details for Web of Science ID 000222650500014

    View details for PubMedID 15238523

  • Genetic dissection of meiotic cytokinesis in Drosophila males MOLECULAR BIOLOGY OF THE CELL Giansanti, M. G., Farkas, R. M., Bonaccorsi, S., Lindsley, D. L., Wakimoto, B. T., Fuller, M. T., Gatti, M. 2004; 15 (5): 2509-2522

    Abstract

    We have used Drosophila male meiosis as a model system for genetic dissection of the cytokinesis mechanism. Drosophila mutants defective in meiotic cytokinesis can be easily identified by their multinucleate spermatids. Moreover, the large size of meiotic spindles allows characterization of mutant phenotypes with exquisite cytological resolution. We have screened a collection of 1955 homozygous mutant male sterile lines for those with multinucleate spermatids, and thereby identified mutations in 19 genes required for cytokinesis. These include 16 novel loci and three genes, diaphanous, four wheel drive, and pebble, already known to be involved in Drosophila cytokinesis. To define the primary defects leading to failure of cytokinesis, we analyzed meiotic divisions in male mutants for each of these 19 genes. Examination of preparations stained for tubulin, anillin, KLP3A, and F-actin revealed discrete defects in the components of the cytokinetic apparatus, suggesting that these genes act at four major points in a stepwise pathway for cytokinesis. Our results also indicated that the central spindle and the contractile ring are interdependent structures that interact throughout cytokinesis. Moreover, our genetic and cytological analyses provide further evidence for a cell type-specific control of Drosophila cytokinesis, suggesting that several genes required for meiotic cytokinesis in males are not required for mitotic cytokinesis.

    View details for Web of Science ID 000221189300039

    View details for PubMedID 15004238

  • Regulation of transcription of meiotic cell cycle and terminal differentiation genes by the testis-specific Zn-finger protein matotopetli DEVELOPMENT Perezgasga, L., Jiang, J. Q., Bolival, B., Hiller, M., Benson, E., Fuller, M. T., White-Cooper, H. 2004; 131 (8): 1691-1702

    Abstract

    A robust developmentally regulated and cell type specific transcriptional programme is activated in primary spermatocytes in preparation for differentiation of the male gametes during spermatogenesis. Work in Drosophila is beginning to reveal the genetic networks that regulate this gene expression. The Drosophila aly-class meiotic arrest loci are essential for activation of transcription of many differentiation-specific genes, as well as several genes important for meiotic cell cycle progression, thus linking meiotic cell cycle progression to cellular differentiation during spermatogenesis. The three previously described aly-class proteins (aly, comr and achi/vis) form a complex and are associated with chromatin in primary spermatocytes. We identify, clone and characterize a new aly-class meiotic arrest gene, matotopetli (topi), which encodes a testis-specific Zn-finger protein that physically interacts with Comr. The topi mutant phenotype is most like achi/vis in that topi function is not required for the nuclear localization of Aly or Comr, but is required for their accumulation on chromatin. Most target genes in the transcriptional programme depend on both topi and achi/vis; however, a small subset of target genes are differentially sensitive to loss of topi or achi/vis, suggesting that these aly-class predicted DNA binding proteins can act independently in some contexts.

    View details for DOI 10.1242/dev.01032

    View details for Web of Science ID 000221155900003

    View details for PubMedID 15084455

  • Germ line stem cell differentiation in Drosophila requires gap junctions and proceeds via an intermediate state DEVELOPMENT Gilboa, L., Forbes, A., Tazuke, S. I., Fuller, M. T., Lehmann, R. 2003; 130 (26): 6625-6634

    Abstract

    Gap junctions coordinate processes ranging from muscle contraction to ovarian follicle development. Here we show that the gap junction protein Zero population growth (Zpg) is required for germ cell differentiation in the Drosophila ovary. In the absence of Zpg the stem cell daughter destined to differentiate dies. The zpg phenotype is novel, and we used this phenotype to genetically dissect the process of stem cell maintenance and differentiation. Our findings suggest that germ line stem cells differentiate upon losing contact with their niche, that gap junction mediated cell-cell interactions are required for germ cell differentiation, and that in Drosophila germ line stem cell differentiation to a cystoblast is gradual.

    View details for DOI 10.1242/dev.00853

    View details for Web of Science ID 000188254100021

    View details for PubMedID 14660550

  • Mitofusin-1 protein is a generally expressed mediator of mitochondrial fusion in mammalian cells JOURNAL OF CELL SCIENCE Santel, A., Frank, S., Gaume, B., Herrier, M., Youle, R. J., Fuller, M. T. 2003; 116 (13): 2763-2774

    Abstract

    Mitochondrial fusion may regulate mitochondrial morphogenesis and underlie complementation between mitochondrial genomes in mammalian cells. The nuclear encoded mitochondrial proteins Mfn1 and Mfn2 are human homologues of the only known protein mediators of mitochondrial fusion, the Drosophila Fzo GTPase and Saccharomyces cerevisiae yFzo1p. Although the Mfn1 and Mfn2 genes were broadly expressed, the two genes showed different levels of mRNA expression in different tissues. Two Mfn1 transcripts were detected at similar levels in a variety of human tissues and were dramatically elevated in heart, while Mfn2 mRNA was abundantly expressed in heart and muscle tissue but present only at low levels in many other tissues. Human Mfn1 protein localized to mitochondria and participated in a high molecular weight, detergent extractable protein complex. Forced expression of Mfn1 in cultured cells caused formation of characteristic networks of mitochondria. Introduction of a point mutation in the conserved G1 region of the predicted GTPase domain (Mfn1K88T) dramatically decreased formation of mitochondrial networks upon Mfn1 overexpression, suggesting that network formation required completion of the Mfn1 GTPase cycle. Conversely, a protein variant carrying a point mutation in the G2 motif of the Mfn1 GTPase domain acted as a dominant negative: overexpression of Mfn1T109A resulted in fragmentation of mitochondria. We propose that Mfn1T109A interferes with fusion activity of endogenous Mfn1 protein, possibly by binding necessary cofactors, so to allow unopposed mitochondrial fission. Thus, Mfn1 appears to be a key player in mediating mitochondrial fusion and morphology in mammalian cells.

    View details for DOI 10.1242/jcs.00479

    View details for Web of Science ID 000184096800015

    View details for PubMedID 12759376

  • The Drosophila Cog5 homologue is required for cytokinesis, cell elongation, and assembly of specialized golgi architecture during spermatogenesis MOLECULAR BIOLOGY OF THE CELL Farkas, R. M., Giansanti, M. G., Gatti, M., Fuller, M. T. 2003; 14 (1): 190-200

    Abstract

    The multisubunit conserved oligomeric Golgi (COG) complex has been shown previously to be involved in Golgi function in yeast and mammalian tissue culture cells. Despite this broad conservation, several subunits, including Cog5, were not essential for growth and showed only mild effects on secretion when mutated in yeast, raising questions about what functions these COG complex subunits play in the life of the cell. Here, we show that function of the gene four way stop (fws), which encodes the Drosophila Cog5 homologue, is necessary for dramatic changes in cellular and subcellular morphology during spermatogenesis. Loss-of-function mutations in fws caused failure of cleavage furrow ingression in dividing spermatocytes and failure of cell elongation in differentiating spermatids and disrupted the formation and/or stability of the Golgi-based spermatid acroblast. Consistent with the lack of a growth defect in yeast lacking Cog5, animals lacking fws function were viable, although males were sterile. Fws protein localized to Golgi structures throughout spermatogenesis. We propose that Fws may directly or indirectly facilitate efficient vesicle traffic through the Golgi to support rapid and extensive increases in cell surface area during spermatocyte cytokinesis and polarized elongation of differentiating spermatids. Our study suggests that Drosophila spermatogenesis can be an effective sensitized genetic system to uncover in vivo functions for proteins involved in Golgi architecture and/or vesicle transport.

    View details for DOI 10.1091/mbc.E02-06-0343

    View details for Web of Science ID 000180497300016

    View details for PubMedID 12529436

    View details for PubMedCentralID PMC140237

  • Spatial and temporal association of Bax with mitochondrial fission sites, Drp1, and Mfn2 during apoptosis JOURNAL OF CELL BIOLOGY Karbowski, M., Lee, Y. J., Gaume, B., Jeong, S. Y., Frank, S., Nechushtan, A., Santel, A., Fuller, M., Smith, C. L., Youle, R. J. 2002; 159 (6): 931-938

    Abstract

    We find that Bax, a proapoptotic member of the Bcl-2 family, translocates to discrete foci on mitochondria during the initial stages of apoptosis, which subsequently become mitochondrial scission sites. A dominant negative mutant of Drp1, Drp1K38A, inhibits apoptotic scission of mitochondria, but does not inhibit Bax translocation or coalescence into foci. However, Drp1K38A causes the accumulation of mitochondrial fission intermediates that are associated with clusters of Bax. Surprisingly, Drp1 and Mfn2, but not other proteins implicated in the regulation of mitochondrial morphology, colocalize with Bax in these foci. We suggest that Bax participates in apoptotic fragmentation of mitochondria.

    View details for DOI 10.1083/jcb.200209124

    View details for Web of Science ID 000180150200003

    View details for PubMedID 12499352

    View details for PubMedCentralID PMC2173996

  • Signaling from germ cells mediated by the rhomboid homolog stet organizes encapsulation by somatic support cells DEVELOPMENT Schulz, C., Wood, C. G., Jones, D. L., Tazuke, S. I., Fuller, M. T. 2002; 129 (19): 4523-4534

    Abstract

    Germ cells normally differentiate in the context of encapsulating somatic cells. However, the mechanisms that set up the special relationship between germ cells and somatic support cells and the signals that mediate the crucial communications between the two cell types are poorly understood. We show that interactions between germ cells and somatic support cells in Drosophila depend on wild-type function of the stet gene. In males, stet acts in germ cells to allow their encapsulation by somatic cyst cells and is required for germ cell differentiation. In females, stet function allows inner sheath cells to enclose early germ cells correctly at the tip of the germarium. stet encodes a homolog of rhomboid, a component of the epidermal growth factor receptor signaling pathway involved in ligand activation in the signaling cell. The stet mutant phenotype suggests that stet facilitates signaling from germ cells to the epidermal growth factor receptor on somatic cells, resulting in the encapsulation of germ cells by somatic support cells. The micro-environment provided by the surrounding somatic cells may, in turn, regulate differentiation of the germ cells they enclose.

    View details for Web of Science ID 000178640100013

    View details for PubMedID 12223409

  • Differential expression of the Drosophila mitofusin genes fuzzy onions (fzo) and dmfn MECHANISMS OF DEVELOPMENT Hwa, J. J., Hiller, M. A., Fuller, M. T., Santel, A. 2002; 116 (1-2): 213-216

    Abstract

    Mitofusins comprise a family of evolutionarily conserved, nuclear encoded mitochondrial guanosine triphoshatases that control mitochondrial fusion and morphology. The fuzzy onions (fzo) and Drosophila mitofusin (dmfn) genes, which encode the only Mitofusin homologs in Drosophila are differentially expressed during development. Dmfn-mRNA was widely expressed during embryogenesis accumulating in the mesoderm and endoderm during gut development, during oogenesis with transcripts maternally deposited into the early embryo and in the male germ line, where dmfn-mRNA was expressed in spermatogonia, spermatocytes and early spermatids. In contrast, expression of the fzo was tightly restricted to the male germ line, with mRNA accumulation in spermatocytes and early spermatids. In addition, expression of dmfn and fzo in the same cell type, primary spermatocytes, was under control of different regulatory mechanisms.

    View details for Web of Science ID 000177333000027

    View details for PubMedID 12128227

  • A germline-specific gap junction protein required for survival of differentiating early germ cells DEVELOPMENT Tazuke, S. I., Schulz, C., Gilboa, L., Fogarty, M., Mahowald, A. P., Guichet, A., Ephrussi, A., Wood, C. G., Lehmann, R., Fuller, M. T. 2002; 129 (10): 2529-2539

    Abstract

    Germ cells require intimate associations and signals from the surrounding somatic cells throughout gametogenesis. The zero population growth (zpg) locus of Drosophila encodes a germline-specific gap junction protein, Innexin 4, that is required for survival of differentiating early germ cells during gametogenesis in both sexes. Animals with a null mutation in zpg are viable but sterile and have tiny gonads. Adult zpg-null gonads contain small numbers of early germ cells, resembling stem cells or early spermatogonia or oogonia, but lack later stages of germ cell differentiation. In the male, Zpg protein localizes to the surface of spermatogonia, primarily on the sides adjacent to the somatic cyst cells. In the female, Zpg protein localizes to germ cell surfaces, both those adjacent to surrounding somatic cells and those adjacent to other germ cells. We propose that Zpg-containing gap junctional hemichannels in the germ cell plasma membrane may connect with hemichannels made of other innexin isoforms on adjacent somatic cells. Gap junctional intercellular communication via these channels may mediate passage of crucial small molecules or signals between germline and somatic support cells required for survival and differentiation of early germ cells in both sexes.

    View details for Web of Science ID 000176063600019

    View details for PubMedID 11973283

  • Stem cell self-renewal specified by JAK-STAT activation in response to a support cell cue SCIENCE Kiger, A. A., Jones, D. L., Schulz, C., Rogers, M. B., Fuller, M. T. 2001; 294 (5551): 2542-2545

    Abstract

    Stem cells generate many differentiated, short-lived cell types, such as blood, skin, and sperm, throughout adult life. Stem cells maintain a long-term capacity to divide, producing daughter cells that either self-renew or initiate differentiation. Although the surrounding microenvironment or "niche" influences stem cell fate decisions, few signals that emanate from the niche to specify stem cell self-renewal have been identified. Here we demonstrate that the apical hub cells in the Drosophila testis act as a cellular niche that supports stem cell self-renewal. Hub cells express the ligand Unpaired (Upd), which activates the Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway in adjacent germ cells to specify self-renewal and continual maintenance of the germ line stem cell population.

    View details for Web of Science ID 000172927700057

    View details for PubMedID 11752574

  • Genetic analysis of dPsa, the Drosophila orthologue of puromycin-sensitive aminopeptidase, suggests redundancy of aminopeptidases DEVELOPMENT GENES AND EVOLUTION Schulz, C., Perezgasga, L., Fuller, M. T. 2001; 211 (12): 581-588

    Abstract

    Abstract. The Drosophila genome contains a single orthologue of mammalian puromycin-sensitive aminopeptidases, dPsa. Even though dPsa was expressed in many tissues during development, animals lacking dPsa activity were viable. Ubiquitous overexpression of dPsa during embryonic or larval development resulted in lethality and overexpression in isolated tissues during development resulted in localized lesions. These results suggest that even though dPsa function was not essential for viability, dPsa expression must be tightly regulated for normal development. By screening the Drosophila genome we found 43 predicted aminopeptidases and generated a phylogenetic tree of aminopeptidases related to dPsa by sequence. We discuss possible functions of dPsa and the idea that other Drosophila aminopeptidases might perform redundant functions with dPsa for regulating protein turnover.

    View details for DOI 10.1007/s00427-001-0194-z

    View details for Web of Science ID 000174249000002

    View details for PubMedID 11819115

  • Developmental regulation of transcription by a tissue-specific TAF homolog GENES & DEVELOPMENT Hiller, M. A., Lin, T. Y., Wood, C., Fuller, M. T. 2001; 15 (8): 1021-1030

    Abstract

    Alternate forms of the general transcription machinery have been described in several tissues or cell types. However, the role of tissue-specific TBP-associated factors (TAF(II)s) and other tissue-specific transcription components in regulating differential gene expression during development was not clear. Here we show that the cannonball gene of Drosophila encodes a cell type-specific homolog of a more ubiquitously expressed component of the general transcription factor TFIID. cannonball is required in vivo for high level transcription of a set of stage- and tissue-specific target genes during male gametogenesis. Regulation of transcription by cannonball is absolutely required for spermatogenesis, as null mutations block meiotic cell cycle progression and result in a complete failure of spermatid differentiation. Our results demonstrate that cell type-specific TAF(II)s play an important role in developmental regulation of gene expression.

    View details for Web of Science ID 000168271800010

    View details for PubMedID 11316795

  • Control of mitochondrial morphology by a human mitofusin JOURNAL OF CELL SCIENCE Santel, A., Fuller, M. T. 2001; 114 (5): 867-874

    Abstract

    Although changes in mitochondrial size and arrangement accompany both cellular differentiation and human disease, the mechanisms that mediate mitochondrial fusion, fission and morphogenesis in mammalian cells are not understood. We have identified two human genes encoding potential mediators of mitochondrial fusion. The mitofusins (Mfn1 and Mfn2) are homologs of the Drosophila protein fuzzy onion (Fzo) that associate with mitochondria and alter mitochondrial morphology when expressed by transient transfection in tissue culture cells. An internal region including a predicted bipartite transmembrane domain (TM) is sufficient to target Mfn2 to mitochondria and requires hydrophobic residues within the TM. Co-expression of Mfn2 with a dominant interfering mutant dynamin-related protein (Drp1(K38A)) proposed to block mitochondrial fission resulted in long mitochondrial filaments and networks. Formation of mitochondrial filaments and networks required a wild-type Mfn2 GTPase domain, suggesting that the Mfn2 GTPase regulates or mediates mitochondrial fusion and that mitofusins and dynamin related GTPases play opposing roles in mitochondrial fusion and fission in mammals, as in yeast.

    View details for Web of Science ID 000167569000005

    View details for PubMedID 11181170

  • Transcription of meiotic cell cycle and terminal differentiation genes depends on a conserved chromatin associated protein, whose nuclear localisation is regulated DEVELOPMENT White-Cooper, H., Leroy, D., MacQueen, A., Fuller, M. T. 2000; 127 (24): 5463-5473

    Abstract

    The Drosophila always early (aly) gene coordinately regulates meiotic cell cycle progression and terminal differentiation during male gametogenesis. aly is required for transcription of key G2-M cell cycle control genes and of spermatid differentiation genes, and for maintenance of normal chromatin structure in primary spermatocytes. We show that aly encodes a homologue of the Caenorhabditis elegans gene lin-9, a negative regulator of vulval development that acts in the same SynMuvB genetic pathway as the LIN-35 Rb-like protein. The aly gene family is conserved from plants to humans. Aly protein is both cytoplasmic and nuclear in early primary spermatocytes, then resolves to a chromatin-associated pattern. It remains cytoplasmic in a loss-of-function missense allele, suggesting that nuclear localisation is critical for Aly function, and that other factors may alter Aly activity by controlling its subcellular localisation. MAPK activation occurs normally in aly mutant testes. Therefore aly, and by inference lin-9, act in parallel to, or downstream of, activation of MAPK by the RTK-Ras signalling pathway. We favour a model where aly may regulate cell cycle progression and terminal differentiation during male gametogenesis by regulating chromatin conformation in primary spermatocytes.

    View details for Web of Science ID 000166204800022

    View details for PubMedID 11076766

  • Somatic support cells restrict germline stem cell self-renewal and promote differentiation NATURE Kiger, A. A., White-Cooper, H., Fuller, M. T. 2000; 407 (6805): 750-754

    Abstract

    Stem cells maintain populations of highly differentiated, short-lived cell-types, including blood, skin and sperm, throughout adult life. Understanding the mechanisms that regulate stem cell behaviour is crucial for realizing their potential in regenerative medicine. A fundamental characteristic of stem cells is their capacity for asymmetric division: daughter cells either retain stem cell identity or initiate differentiation. However, stem cells are also capable of symmetric division where both daughters remain stem cells, indicating that mechanisms must exist to balance self-renewal capacity with differentiation. Here we present evidence that support cells surrounding the stem cells restrict self-renewal and control stem cell number by ensuring asymmetric division. Loss of function of the Drosophila Epidermal growth factor receptor in somatic cells disrupted the balance of self-renewal versus differentiation in the male germline, increasing the number of germline stem cells. We propose that activation of this receptor specifies normal behaviour of somatic support cells; in turn, the somatic cells play a guardian role, providing information that prevents self-renewal of stem cell identity by the germ cell they enclose.

    View details for Web of Science ID 000089773900044

    View details for PubMedID 11048722

  • A phospholipid kinase regulates actin organization and intercellular bridge formation during germline cytokinesis DEVELOPMENT Brill, J. A., Hime, G. R., Scharer-Schuksz, M., Fuller, M. T. 2000; 127 (17): 3855-3864

    Abstract

    The endgame of cytokinesis can follow one of two pathways depending on developmental context: resolution into separate cells or formation of a stable intercellular bridge. Here we show that the four wheel drive (fwd) gene of Drosophila melanogaster is required for intercellular bridge formation during cytokinesis in male meiosis. In fwd mutant males, contractile rings form and constrict in dividing spermatocytes, but cleavage furrows are unstable and daughter cells fuse together, producing multinucleate spermatids. fwd is shown to encode a phosphatidylinositol 4-kinase (PI 4-kinase), a member of a family of proteins that perform the first step in the synthesis of the key regulatory membrane phospholipid PIP2. Wild-type activity of the fwd PI 4-kinase is required for tyrosine phosphorylation in the cleavage furrow and for normal organization of actin filaments in the constricting contractile ring. Our results suggest a critical role for PI 4-kinases and phosphatidylinositol derivatives during the final stages of cytokinesis.

    View details for Web of Science ID 000089430700020

    View details for PubMedID 10934029

  • Regulation of meiosis and spermatid differentiation in Drosophila primary spermatocytes XVth Testis Workshop on the Testis - From Stem Cell to Sperm Function Lin, T. Y., Pringle, M. J., Fuller, M. T. SPRINGER-VERLAG. 2000: 120–132
  • Developmental genetics of the essential Drosophila nucleoporin nup154: Allelic differences due to an outward-directed promoter in the P-element 3 ' end GENETICS Kiger, A. A., Gigliotti, S., Fuller, M. T. 1999; 153 (2): 799-812

    Abstract

    Drosophila nup154 encodes a predicted nucleoporin homologous to yeast Nup170p, Nup157p, and vertebrate Nup155, all of which are major components of the nuclear pore complex (NPC). Unlike its yeast homologs, nup154 is essential for viability. Animals with strong loss-of-function nup154 mutations caused by P-element insertion in the 5'-UTR of the gene died as larvae with small discs, brains, and testes. nup154 mRNA expression appeared developmentally regulated in tissues of wild-type embryos, larvae, and adults, suggesting that new nup154 synthesis is required when assembly of new NPCs is required, as in proliferating or growing tissues. Two additional nup154 alleles also associated with different P-element inserts in the 5'-UTR were viable but had strong loss-of-function sterile phenotypes, including failure to maintain spermatogenic stem cells and failure to progress into vitellogenic stages of oogenesis. Lethality vs. viability correlated with orientation of the P-element inserts in the different alleles. Transcript analysis by 5'-RACE suggested a mechanism for allelic differences: an outward-directed promoter internal to the P-element 3' end able to drive sufficient expression of the nup154 transcript for viability but not for fertility.

    View details for Web of Science ID 000083051400024

    View details for PubMedID 10511559

  • Molecular characterization of mutant alleles of the DNA repair basal transcription factor haywire/ERCC3 in Drosophila GENETICS Mounkes, L. C., Fuller, M. T. 1999; 152 (1): 291-297

    Abstract

    The haywire gene of Drosophila encodes a putative helicase essential for transcription and nucleotide excision repair. A haywire allele encoding a dominant acting poison product, lethal alleles, and viable but UV-sensitive alleles isolated as revertants of the dominant acting poison allele were molecularly characterized. Sequence analysis of lethal haywire alleles revealed the importance of the nucleotide-binding domain, suggesting an essential role for ATPase activity. The viable haync2 allele, which encodes a poison product, has a single amino acid change in conserved helicase domain VI. This mutation results in accumulation of a 68-kD polypeptide that is much more abundant than the wild-type haywire protein.

    View details for Web of Science ID 000080219100024

    View details for PubMedID 10224261

  • Mitochondrial fusion in yeast requires the transmembrane GTPase Fzo1p. Hermann, G. J., Thatcher, J. W., Mills, J. P., Hales, K. G., Fuller, M. T., Nunnari, J., Shaw, J. M. CELL PRESS. 1999: A2–A2
  • Mitochondrial fusion in yeast requires the transmembrane GTPase Fzo1p JOURNAL OF CELL BIOLOGY Hermann, G. J., Thatcher, J. W., Mills, J. P., Hales, K. G., Fuller, M. T., Nunnari, J., Shaw, J. M. 1998; 143 (2): 359-373

    Abstract

    Membrane fusion is required to establish the morphology and cellular distribution of the mitochondrial compartment. In Drosophila, mutations in the fuzzy onions (fzo) GTPase block a developmentally regulated mitochondrial fusion event during spermatogenesis. Here we report that the yeast orthologue of fuzzy onions, Fzo1p, plays a direct and conserved role in mitochondrial fusion. A conditional fzo1 mutation causes the mitochondrial reticulum to fragment and blocks mitochondrial fusion during yeast mating. Fzo1p is a mitochondrial integral membrane protein with its GTPase domain exposed to the cytoplasm. Point mutations that alter conserved residues in the GTPase domain do not affect Fzo1p localization but disrupt mitochondrial fusion. Suborganellar fractionation suggests that Fzo1p spans the outer and is tightly associated with the inner mitochondrial membrane. This topology may be required to coordinate the behavior of the two mitochondrial membranes during the fusion reaction. We propose that the fuzzy onions family of transmembrane GTPases act as molecular switches to regulate a key step in mitochondrial membrane docking and/or fusion.

    View details for Web of Science ID 000076618200008

    View details for PubMedID 9786948

    View details for PubMedCentralID PMC2132826

  • Genetic control of cell proliferation and differentiation in Drosophila spermatogenesis SEMINARS IN CELL & DEVELOPMENTAL BIOLOGY Fuller, M. T. 1998; 9 (4): 433-444

    Abstract

    Outlines of the genetic circuitry regulating male gametogenesis in Drosophila have begun to appear. Cessation of mitotic proliferation and onset of the meiotic program is regulated by the bam and bgcn genes acting within male germ cells and a TGF-beta class signaling cascade in surrounding somatic cells. Onset of spermatid differentiation is regulated by a stage- and tissue-specific transcriptional program controlled by the aly, can, mia and sa genes. A cross-regulatory mechanism might act, in part by controlling expression of the twine cell cycle phosphatase, to delay the G2/M transition of meiosis I until genes required for spermatid differentiation have been transcribed.

    View details for Web of Science ID 000077088400007

    View details for PubMedID 9813190

  • The zero population growth (zpg) gene is required for mitotic amplification stage of early germ cells in Drosophila. Tazuke, S., Schulz, C., Fogarty, M., Wood, C., Guichet, A., Ephrussi, A., Fuller, M. T. ACADEMIC PRESS INC ELSEVIER SCIENCE. 1998: 197–97
  • The DUG gene of Drosophila melanogaster encodes a structural and functional homolog of the S-cerevisiae SUG1 predicted ATPase associated with the 26S proteasome GENE Mounkes, L. C., Fuller, M. T. 1998; 206 (2): 165-174

    Abstract

    The DUG gene of Drosophila encodes a putative ATPase that is a structural and functional homolog of the yeast SUG1 product. When introduced into S. cerevisiae, the Drosophila DUG gene rescued the lethality associated with a SUG1 mutant. Anti-DUG antibodies recognized a protein that migrated in high molecular weight complexes, along with components of the 26S proteasome, and also immunoprecipitated components of the 26S proteasome from embryonic extracts. Proteins recognized by the affinity-purified antibody raised against DUG were localized in either a punctate cytoplasmic distribution or in the nucleus, depending on the cell type, consistent with the subcellular localization of the 26S proteasome in various cell types.

    View details for Web of Science ID 000072104800002

    View details for PubMedID 9469929

  • Transcriptional and post-transcriptional control mechanisms coordinate the onset of spermatid differentiation with meiosis I in Drosophila DEVELOPMENT White-Cooper, H., Schafer, M. A., Alphey, L. S., Fuller, M. T. 1998; 125 (1): 125-134

    Abstract

    The aly, can, mia and sa genes of Drosophila are essential in males both for the G2-meiosis I transition and for onset of spermatid differentiation. Function of all four genes is required for transcription in primary spermatocytes of a suite of spermatid differentiation genes. aly is also required for transcription of the cell cycle control genes cyclin B and twine in primary spermatocytes. In contrast can, mia and sa are required for accumulation of twine protein but not twine transcript. We propose that the can, mia and sa gene products act together or in a pathway to turn on transcription of spermatid differentiation genes, and that aly acts upstream of can, mia and sa to regulate spermatid differentiation. We also propose that control of translation or protein stability regulates entry into the first meiotic division. We suggest that a gene or genes transcribed under the control of can, mia and sa allow(s) accumulation of twine protein, thus coordinating meiotic division with onset of spermatid differentiation.

    View details for Web of Science ID 000071652400013

    View details for PubMedID 9389670

  • A chromatin-associated kinesin-related protein required for normal mitotic chromosome segregation in Drosophila JOURNAL OF CELL BIOLOGY Molina, I., Baars, S., Brill, J. A., Hales, K. G., Fuller, M. T., Ripoll, P. 1997; 139 (6): 1361-1371

    Abstract

    The tiovivo (tio) gene of Drosophila encodes a kinesin-related protein, KLP38B, that colocalizes with condensed chromatin during cell division. Wild-type function of the tio gene product KLP38B is required for normal chromosome segregation during mitosis. Mitotic cells in tio larval brains displayed circular mitotic figures, increased ploidy, and abnormal anaphase figures. KLP38B mRNA is maternally provided and expressed in cells about to undergo division. We propose that KLP38B, perhaps redundantly with other chromosome-associated microtubule motor proteins, contributes to interactions between chromosome arms and microtubules important for establishing bipolar attachment of chromosomes and assembly of stable bipolar spindles.

    View details for Web of Science ID 000071147400001

    View details for PubMedID 9396743

  • Developmentally regulated mitochondrial fusion mediated by a conserved, novel, predicted GTPase CELL Hales, K. G., Fuller, M. T. 1997; 90 (1): 121-129

    Abstract

    The Drosophila melanogaster fuzzy onions (fzo) gene encodes the first known protein mediator of mitochondrial fusion. During Drosophila spermatogenesis, mitochondria in early postmeiotic spermatids aggregate, fuse, and elongate beside the growing flagellar axoneme. fzo mutant males are defective in this developmentally regulated mitochondrial fusion and are sterile. fzo encodes a large, novel, predicted transmembrane GTPase that becomes detectable on spermatid mitochondria late in meiosis II, just prior to fusion, and disappears soon after fusion is complete. Missense mutations that alter conserved residues required for GTP binding in other GTPases inhibit the fusogenic activity of Fzo in vivo but do not affect its localization. Fzo has homologs of unknown function in mammals, nematodes, and yeast.

    View details for Web of Science ID A1997XL36200014

    View details for PubMedID 9230308

  • Differential expression of two gamma-tubulin isoforms during gametogenesis and development in Drosophila DEVELOPMENTAL BIOLOGY Wilson, P. G., Zheng, Y., Oakley, C. E., Oakley, B. R., Borisy, G. G., Fuller, M. T. 1997; 184 (2): 207-221

    Abstract

    Previous work identified a gamma-tubulin gene, gamma Tub23C, in Drosophila (Zheng et al., 1991). We now report identification of a second gamma-tubulin gene, gamma Tub37CD. Immunoblot analysis and immunolocalization show that gamma Tub37CD and gamma Tub23C are differentially expressed during gametogenesis and development. During oogenesis, gamma Tub23C was detected at centrosomes and in the cytoplasm of mitotic germ cells, but was not detected in germ cells following completion of mitosis. Conversely, gamma Tub37CD was not detected in proliferating germ cells, but appeared to accumulate in germ cells during egg chamber development. Neither gamma-tubulin isoform was detected at the anterior or posterior poles of developing oocytes. During spermatogenesis, only gamma Tub23C was detected at centrosomes, where it showed cell cycle- and differentiation-dependent organization. During the transition into the first meiotic division, gamma Tub23C became organized as a corpuscular focus at centrioles until completion of meiosis II. During postmeiotic spermatid differentiation, gamma Tub23C was detected first as a rod and then as a collar-like structure near the juncture of the nucleus and the elongating flagellum, but was not detected in bundles of mature sperm. The germline-specific CDC25 encoded by twine is required for organization of gamma Tub23C into corpuscular focus in spermatocytes, but not for separation of centriole pairs in M-phase or postmeiotic organization of gamma Tub23C at centrioles. Following reconstitution of a canonical centrosome at fertilization, only gamma Tub37CD was detected at centrosomes in syncytial embryos, but both gamma Tub37CD and gamma Tub23C were detected at centrosomes in cellularized embryos. Colocalization of these two isoforms suggests that gamma Tub23C and gamma Tub37CD both contain structural features of gamma-tubulins essential for localization to centrosomes.

    View details for Web of Science ID A1997WX47200003

    View details for PubMedID 9133431

  • Monastral bipolar spindles: Implications for dynamic centrosome organization JOURNAL OF CELL SCIENCE Wilson, P. G., Fuller, M. T., Borisy, G. G. 1997; 110: 451-464

    Abstract

    Implicit to all models for mitotic spindle assembly is the view that centrosomes are essentially permanent structures. Yet, immunofluorescence revealed that spindles in larval brains of urchin mutants in Drosophila were frequently monastral but bipolar; the astral pole contained a centrosome while the opposing anastral pole showed neither gamma tubulin nor a radial array of astral microtubules. Thus, mutations in the urchin gene seem to uncouple centrosome organization and spindle bipolarity in mitotic cells. Hypomorphic mutants showed a high frequency of monastral bipolar spindles but low frequencies of polyploidy, suggesting that monastral bipolar spindles might be functional. To test this hypothesis, we performed pedigree analysis of centrosome distribution and spindle structure in the four mitotic divisions of gonial cells. Prophase gonial cells showed two centrosomes, suggesting cells entered mitosis with the normal number of centrosomes and that centrosomes separated during prophase. Despite a high frequency of monastral bipolar spindles, the end products of the four mitotic divisions were equivalent in size and chromatin content. These results indicate that monastral bipolar spindles are functional and that the daughter cell derived from the anastral pole can assemble a functional bipolar spindle in the subsequent cell cycle. Cell proliferation despite high frequencies of monastral bipolar spindles can be explained if centrosome structure in mitotic cells is dynamic, allowing transient and benign disorganization of pericentriolar components. Since urchin proved to be allelic to KLP61F which encodes a kinesin related motor protein (Heck et al. (1993) J. Cell Biol. 123, 665-671), our results suggest that motors influence the dynamic organization of centrosomes.

    View details for Web of Science ID A1997WL46000006

    View details for PubMedID 9067597

  • Assembly of ring canals in the male germ line from structural components of the contractile ring JOURNAL OF CELL SCIENCE Hime, G. R., Brill, J. A., Fuller, M. T. 1996; 109: 2779-2788

    Abstract

    Stable intercellular bridges called ring canals form following incomplete cytokinesis, and interconnect mitotically or meiotically related germ cells. We show that ring canals in Drosophila melanogaster males are surprisingly different from those previously described in females. Mature ring canal walls in males lack actin and appear to derive directly from structural proteins associated with the contractile ring. Ring canal assembly in males, as in females, initiates during cytokinesis with the appearance of a ring of phosphotyrosine epitopes at the site of the contractile ring. Following constriction, actin and myosin II disappear. However, at least four proteins present at the contractile ring remain: the three septins (Pnut, Sep1 and Sep2) and anillin. In sharp contrast, in ovarian ring canals, septins have not been detected, anillin is lost from mature ring canals and filamentous actin is a major component. In both males and females, a highly branched vesicular structure, termed the fusome, interconnects developing germ cells via the ring canals and is thought to coordinate mitotic germ cell divisions. We show that, in males, unlike females, the fusome persists and enlarges following cessation of the mitotic divisions, developing additional branches during meiosis. During differentiation, the fusome and its associated ring canals localize to the distal tip of the elongating spermatids.

    View details for Web of Science ID A1996WB43700001

    View details for PubMedID 9013326

  • Coordinate developmental control of the meiotic cell cycle and spermatid differentiation in Drosophila males DEVELOPMENT Lin, T. Y., Viswanathan, S., Wood, C., Wilson, P. G., Wolf, N., Fuller, M. T. 1996; 122 (4): 1331-1341

    Abstract

    Wild-type function of four Drosophila genes, spermatocyte arrest, cannonball, always early and meiosis I arrest, is required both for cell-cycle progression through the G2/M transition of meiosis I in males and for onset of spermatid differentiation. In males mutant for any one of these meiotic arrest genes, mature primary spermatocytes with partially condensed chromosomes accumulate and postmeiotic cells are lacking. The arrest in cell-cycle progression occurs prior to degradation of cyclin A protein. The block in spermatogenesis in these mutants is not simply a secondary consequence of meiotic cell-cycle arrest, as spermatid differentiation proceeds in males mutant for the cell cycle activating phosphatase twine. Instead, the arrest of both meiosis and spermiogenesis suggests a control point that may serve to coordinate the male meiotic cell cycle with the spermatid differentiation program. The phenotype of the Drosophila meiotic arrest mutants is strikingly similar to the histopathological features of meiosis I maturation arrest infertility in human males, suggesting that the control point may be conserved from flies to man.

    View details for Web of Science ID A1996UF83100029

    View details for PubMedID 8620860

  • RIDING THE POLAR WINDS - CHROMOSOMES MOTOR DOWN EAST CELL Fuller, M. T. 1995; 81 (1): 5-8

    View details for Web of Science ID A1995QR97000002

    View details for PubMedID 7720073

  • A CYTOPLASMIC DYNEIN MOTOR IN DROSOPHILA - IDENTIFICATION AND LOCALIZATION DURING EMBRYOGENESIS JOURNAL OF CELL SCIENCE Hays, T. S., Porter, M. E., McGrail, M., GRISSOM, P., GOSCH, P., Fuller, M. T., McIntosh, J. R. 1994; 107: 1557-1569

    Abstract

    We have characterized a cytoplasmic dynein motor isoform that is present in extracts of Drosophila embryos. A prominent high molecular weight (HMW) polypeptide (> 400 kDa) is enriched in microtubules prepared from nucleotide-depleted embryonic extracts. Based on its ATP-sensitive microtubule binding activity, 20 S sedimentation coefficient, sensitivity to UV-vanadate and nucleotide specificity, the HMW polypeptide resembles cytoplasmic dyneins prepared from other organisms. The Drosophila cytoplasmic dynein acts as a minus-end motor that promotes microtubule translocation in vitro. A polyclonal antibody raised against the dynein heavy chain polypeptide was used to localize the dynein antigen in whole-mount preparations of embryos by immunofluorescence microscopy. These studies show that the dynein motor is associated with microtubules throughout embryogenesis, including mitotic spindle microtubules and microtubules of the embryonic nervous system.

    View details for Web of Science ID A1994NW49500021

    View details for PubMedID 7962198

  • A DROSOPHILA MODEL FOR XERODERMA-PIGMENTOSUM AND COCKAYNES-SYNDROME - HAYWIRE ENCODES THE FLY HOMOLOG OF ERCC3, A HUMAN EXCISION REPAIR GENE CELL Mounkes, L. C., Jones, R. S., Liang, B. C., Gelbart, W., Fuller, M. T. 1992; 71 (6): 925-937

    Abstract

    The haywire gene of Drosophila encodes a protein with 66% identity to the product of the human ERCC3 gene, associated with xeroderma pigmentosum B (XP-B) and Cockayne's syndrome (CS). XP is a human autosomal recessive disease characterized by extreme sensitivity to ultraviolet irradiation and marked susceptibility to skin cancer. In addition, XP and CS patients often exhibit a variety of defects, ranging from central nervous system disorders to hypogonadism. Phenotypes of haywire mutants mimic some of the effects of XP. Many haywire alleles are recessive lethal, viable alleles cause ultraviolet sensitivity, and files expressing marginal levels of haywire display motor defects and reduced life span. Progeny of females carrying a maternal effect allele show central nervous system defects.

    View details for Web of Science ID A1992KB99000006

    View details for PubMedID 1458540

  • FORCE AND COUNTERFORCE IN THE MITOTIC SPINDLE CELL Fuller, M. T., Wilson, P. G. 1992; 71 (4): 547-550

    View details for Web of Science ID A1992JY67600002

    View details for PubMedID 1423613

  • DIFFERENTIAL EXPRESSION AND BEHAVIOR OF 2 GAMMA TUBULIN ISOFORMS IN DROSOPHILA Wilson, P. G., Zheng, Y., Oakley, E., Oakley, B., Fuller, M. T. AMER SOC CELL BIOLOGY. 1992: A346–A346
  • 2 TYPES OF GENETIC INTERACTION IMPLICATE THE WHIRLIGIG GENE OF DROSOPHILA-MELANOGASTER IN MICROTUBULE ORGANIZATION IN THE FLAGELLAR AXONEME GENETICS Green, L. L., Wolf, N., McDonald, K. L., Fuller, M. T. 1990; 126 (4): 961-973

    Abstract

    The mutant nc4 allele of whirligig (3-54.4) of Drosophila melanogaster fails to complement mutations in an alpha-tubulin locus, alpha 1t, mutations in a beta-tubulin locus, B2t, or a mutation in the haywire locus. However, wrl fails to map to any of the known alpha- or beta-tubulin genes. The extragenic failure to complement could indicate that the wrl product participates in structural interactions with microtubule proteins. The whirligig locus appears to be haploinsufficient for male fertility. Both a deficiency of wrl and possible loss of function alleles obtained by reverting the failure to complement between wrlnc4 and B2tn are dominant male sterile in a genetic background wild type for tubulin. The dominant male sterility of the revertant alleles is suppressed if the flies are also heterozygous for B2tn, for a deficiency of alpha 1t, or for the haync2 allele. These results suggest that it is not the absolute level of wrl gene product but its level relative to tubulin or microtubule function that is important for normal spermatogenesis. The phenotype of homozygous wrl mutants suggests that the whirligig product plays a role in postmeiotic spermatid differentiation, possibly in organizing the microtubules of the sperm flagellar axoneme. Flies homozygous for either wrlnc4 or revertant alleles are viable and female fertile but male sterile. Premeiotic and meiotic stages of spermatogenesis appear normal. However, in post-meiotic stages, flagellar axonemes show loss of the accessory microtubule on the B-subfiber of outer doublet microtubules, outer triplet instead of outer doublet microtubules, and missing central pair microtubules.

    View details for Web of Science ID A1990EL33400016

    View details for PubMedID 2127579

  • INTERACTING GENES THAT AFFECT MICROTUBULE FUNCTION IN DROSOPHILA-MELANOGASTER - 2 CLASSES OF MUTATION REVERT THE FAILURE TO COMPLEMENT BETWEEN HAYNC2 AND MUTATIONS IN TUBULIN GENES GENETICS Regan, C. L., Fuller, M. T. 1990; 125 (1): 77-90

    Abstract

    The recessive male sterile mutation haync2 of Drosophila melanogaster fails to complement certain beta 2-tubulin and alpha-tubulin mutations, suggesting that the haywire product plays a role in microtubule function, perhaps as a structural component of microtubules. The genetic interaction appears to require the presence of the aberrant product encoded by haync2, which may act as a structural poison. Based on this observation, we have isolated ten new mutations that revert the failure to complement between haync2 and B2tn. The revertants tested behaved as intragenic mutations of hay in recombination tests, and fell into two phenotypic classes, suggesting two functional domains of the hay gene product. Some revertants were hemizygous viable and less severe than haync2 in their recessive phenotype. These mutations might revert the poison by restoring the aberrant product encoded by the haync2 allele to more wild-type function. Most of the revertants were recessive lethal mutations, indicating that the hay gene product is essential for viability. These more extreme mutations could revert the poison by destroying the ability of the aberrant haywirenc2 product to interact structurally with microtubules. Flies heterozygous for the original haync2 allele and an extreme revertant show defects in both the structure and the function of the male meiotic spindle.

    View details for Web of Science ID A1990DA94700008

    View details for PubMedID 2111265

  • INTERACTING PROTEINS IDENTIFIED BY GENETIC INTERACTIONS - A MISSENSE MUTATION IN ALPHA-TUBULIN FAILS TO COMPLEMENT ALLELES OF THE TESTIS-SPECIFIC BETA-TUBULIN GENE OF DROSOPHILA-MELANOGASTER MOLECULAR AND CELLULAR BIOLOGY Hays, T. S., Deuring, R., Robertson, B., Prout, M., Fuller, M. T. 1989; 9 (3): 875-884

    Abstract

    In this paper we demonstrate that failure to complement between mutations at separate loci can be used to identify genes that encode interacting structural proteins. A mutation (nc33) identified because it failed to complement mutant alleles of the gene encoding the testis-specific beta 2-tubulin of Drosophila melanogaster (B2t) did not map to the B2t locus. We show that this second-site noncomplementing mutation is a missense mutation in alpha-tubulin that results in substitution of methionine in place of valine at amino acid 177. Because alpha- and beta-tubulin form a heterodimer, our results suggest that the genetic interaction, failure to complement, is based on the structural interaction between the protein products of the two genes. Although the nc33 mutation failed to complement a null allele of B2t (B2tn), a deletion of the alpha-tubulin gene to which nc33 mapped complemented B2tn. Thus, the failure to complement appears to require the presence of the altered alpha-tubulin encoded by the nc33 allele, which may act as a structural poison when incorporated into either the tubulin heterodimer or microtubules.

    View details for Web of Science ID A1989T444300001

    View details for PubMedID 2498648

  • INTERACTING GENES IDENTIFY INTERACTING PROTEINS INVOLVED IN MICROTUBULE FUNCTION IN DROSOPHILA CELL MOTILITY AND THE CYTOSKELETON Fuller, M. T., Regan, C. L., Green, L. L., Robertson, B., Deuring, R., Hays, T. S. 1989; 14 (1): 128-135

    View details for Web of Science ID A1989AV59300017

    View details for PubMedID 2684419

  • MUTATIONS THAT ENCODE PARTIALLY FUNCTIONAL BETA-2 TUBULIN SUBUNITS HAVE DIFFERENT EFFECTS ON STRUCTURALLY DIFFERENT MICROTUBULE ARRAYS JOURNAL OF CELL BIOLOGY Fuller, M. T., CAULTON, J. H., Hutchens, J. A., Kaufman, T. C., Raff, E. C. 1988; 107 (1): 141-152

    Abstract

    The testis-specific beta 2 tubulin of Drosophila is required for assembly and function of at least three architecturally different microtubule arrays (Kemphues et al., 1982). Two recessive male-sterile mutations in the B2t locus that encode partially functional, stable, variant forms of beta 2 tubulin cause defects in only certain microtubule-based processes during spermatogenesis. These mutations could thus identify aspects of beta tubulin primary structure critical for function only in specific microtubule arrays. In males carrying the B2t6 mutation, meiotic chromosome segregation and nuclear shaping are normal and flagellar axonemes are formed, but there is a subtle defect in axoneme structure; the outer doublet microtubules fill in with a central core normally seen only in the central pair and accessory microtubules. In homozygous B2t7 males, chromosome movement is usually normal during meiosis but cytokinesis often fails, cytoplasmic microtubules are assembled and nuclear shaping appears to be normal, but the flagellar axoneme lacks structural integrity. In contrast, the B2t8 allele affects a general property of tubulin, the ability to form normal side-to-side association of protofilaments (Fuller et al., 1987), and causes defects in meiosis, axoneme assembly and nuclear shaping. Certain combinations of these beta 2 tubulin mutations show interallelic complementation; in B2t6/B2t8 males functional sperm are produced and both variant subunits are incorporated into mature sperm, in the absence of wild-type beta 2 tubulin. Comparison of the phenotypes of the three partially functional beta 2 tubulin alleles reveals some aspects of tubulin primary structure more important for function in specific subsets of microtubule arrays, and other aspects required for the construction of microtubules in general.

    View details for Web of Science ID A1988P399500014

    View details for PubMedID 3134362

  • TEMPORAL AND SPATIAL PATTERN OF DIFFERENCES IN MICROTUBULE BEHAVIOR DURING DROSOPHILA EMBRYOGENESIS REVEALED BY DISTRIBUTION OF A TUBULIN ISOFORM DEVELOPMENT Wolf, N., Regan, C. L., Fuller, M. T. 1988; 102 (2): 311-324

    Abstract

    Immunofluorescence staining of Drosophila embryos with a monoclonal antibody specific for acetylated alpha-tubulin has revealed that acetylated and nonacetylated alpha-tubulin isoforms have different patterns of distribution during early development. Acetylated alpha-tubulin was not detected in either interphase or mitotic spindle microtubules during the rapid early cleavage or syncytial blastoderm divisions. Acetylated alpha-tubulin was first observed as interphase lengthened at the end of syncytial blastoderm, and at cycle 14 was localized to a ring of structures clustered around the interphase nuclei. These structures probably represent a set of stable microtubules involved in nuclear elongation. Absence of detectable acetylated alpha-tubulin prior to cellular blastoderm seems to be due to rapid turnover of microtubule arrays rather than to lack of the enzyme required for modification, since acetylated alpha-tubulin appeared in early embryos when micro-tubules were stabilized by taxol treatment or anoxia. Because acetylated alpha-tubulin seems to be characteristic of stable microtubule arrays, the appearance of the antigen at cycle 14 represents a fundamental change in microtubule behaviour in the somatic cells of the embryo. Acetylated alpha-tubulin was not detected in pole cells during the blastoderm or early gastrula stages, indicating that acetylation of alpha-tubulin is not merely a consequence of cellularization. After the onset of gastrulation, interphase microtubule arrays in most cell types contain acetylated alpha-tubulin. However, cells in mitosis lack antibody staining. The resulting unstained patches reveal the stereotyped spatial pattern of cell division during gastrulation. Although the cells that give rise to the amnioserosa have acetylated alpha-tubulin in their interphase arrays at early gastrulation, by germ band elongation these large, plastic cells completely lack staining with anti-acetylated alpha-tubulin. In contrast, differentiated cell types such as neurones, which have arrays of stable axonal microtubules, stain brightly with the specific antibody. Although acetylated and nonacetylated alpha-tubulin are present in roughly equal amounts by the late stages of embryogenesis, acetylated alpha-tubulin is partitioned into the pellet during centrifugation of extracts of embryos homogenized at 4 degrees C.

    View details for Web of Science ID A1988M459400007

    View details for PubMedID 3138100

  • INTERACTING GENES THAT AFFECT MICROTUBULE FUNCTION - THE NC2 ALLELE OF THE HAYWIRE LOCUS FAILS TO COMPLEMENT MUTATIONS IN THE TESTIS-SPECIFIC BETA-TUBULIN GENE OF DROSOPHILA GENES & DEVELOPMENT Regan, C. L., Fuller, M. T. 1988; 2 (1): 82-92

    Abstract

    A mutation that fails to complement certain alleles of the testis-specific beta 2-tubulin gene (B2t) of Drosophila melanogaster maps to a separate locus, haywire, located at 3-34.4 map units in polytene region 67E3-F3. Second-site non-complementing mutations such as haync2 and B2t alleles could identify genes that encode products that participate in the same functions or that interact in the same structure. Consistent with a structural interaction between the hay gene product and beta 2-tubulin, the genetic interaction between haync2 and B2t requires the presence of the mutant hay gene product; a deficiency for the hay region complements the same alleles of B2t that haync2 fails to complement. haync2 is a recessive male sterile mutation in a genetic background that is wild type at the B2t locus. Homozygous males have defects in meiosis, flagellar elongation and nuclear shaping, the three major microtubule-based processes in which the testis-specific beta 2-tubulin participates. The haync2 allele also has effects outside of spermatogenesis. It is a temperature-sensitive semilethal mutation, and homozygous haync2 females have reduced fertility. These phenotypes are consistent with a role for the haywire gene product in general microtubule function. Analysis of second-site non-complementing mutations such as haync2 offers a genetic tool for analysis of interacting proteins in complex assemblies.

    View details for Web of Science ID A1988M096400008

    View details for PubMedID 3128461

  • GENETIC-ANALYSIS OF MICROTUBULE STRUCTURE - A BETA-TUBULIN MUTATION CAUSES THE FORMATION OF ABERRANT MICROTUBULES INVIVO AND INVITRO JOURNAL OF CELL BIOLOGY Fuller, M. T., CAULTON, J. H., Hutchens, J. A., Kaufman, T. C., Raff, E. C. 1987; 104 (3): 385-394

    Abstract

    A recessive male sterile mutation (B2t8) that encodes a stable variant of the testis-specific beta 2-tubulin of Drosophila causes the assembly of aberrant microtubules both in vivo and in vitro. The B2t8 mutation appears to cause defects in the formation of interprotofilament bonds. In testes from homozygous mutant males, the most commonly observed aberrant structures were sheets of protofilaments curved to form an S in cross section rather than a normal, closed microtubule. These characteristic S-shaped structures appear in the meiotic spindle, in place of axonemes in differentiating spermatids, and in cytoplasmic microtubules, including those that lie next to the nucleus during nuclear elongation. Homozygous mutant males exhibit defects in chromosome movement and cytokinesis during meiosis, flagellar elongation, and nuclear shaping, indicating that the ability to form normal closed microtubules is required for each of these events. The presence of the aberrant microtubules in three architecturally different microtubule arrays demonstrates conclusively the multifunctional nature of the beta 2-tubulin gene product. Although the mutant beta 2-tubulin subunit causes assembly of aberrant microtubules in vitro and in homozygous males, in the presence of wild-type beta 2-tubulin in heterozygous males, the variant subunit coassembles with the wild-type subunit into functional sperm.

    View details for Web of Science ID A1987G207900002

    View details for PubMedID 3818786

  • ASSEMBLY INVITRO OF BACTERIOPHAGE-P22 PROCAPSIDS FROM PURIFIED COAT AND SCAFFOLDING SUBUNITS JOURNAL OF MOLECULAR BIOLOGY Fuller, M. T., KING, J. 1982; 156 (3): 633-665

    View details for Web of Science ID A1982NN53600012

    View details for PubMedID 6750133

  • STUDIES OF VIRUS STRUCTURE BY LASER RAMAN-SPECTROSCOPY .10. STRUCTURAL STUDIES OF P22 PHAGE, PRECURSOR PARTICLES, AND PROTEINS BY LASER RAMAN-SPECTROSCOPY BIOCHEMISTRY THOMAS, G. J., Li, Y., Fuller, M. T., KING, J. 1982; 21 (16): 3866-3878

    Abstract

    For the study of the protein--protein and protein--nucleic acid interactions in the assembly of virus particles, laser Raman spectra have been obtained in H2O and D2O solutions and as a function of temperature for the following Salmonella phage P22 components: mature phage particles, isolated mature phage DNA, mature protein shells empty of DNA, precursor protein shells (procapsids), and purified coat, scaffolding and tail-spike proteins. The spectra confirm that the condensed DNA within the phage capsid assumes the B-form secondary structure similar to aqueous DNA and reveal no evidence of specific molecular interactions between subgroups of DNA and protein subunits of the phage capsid. No differences were detected in the highly irregular secondary structure of the major capsid protein in mature capsids, empty capsids (lacking DNA), procapsids, and empty procapsids (lacking scaffolding protein). Features of both primary and secondary structures of the viral scaffolding and tail-spike proteins are also revealed by the spectra. Differences in thermal stability of tyrosyl side-chain interactions were observed between scaffolding protein extracted from the procapsid and within the procapsid. These differences correspond to different hydrogen bonding configurations of p-hydroxyphenyl groups and provide indirect evidence for the participation of the scaffolding proteins in specific macromolecular interactions within the procapsid.

    View details for Web of Science ID A1982PA28300023

  • REGULATION OF TUBULIN GENE-EXPRESSION DURING EMBRYOGENESIS IN DROSOPHILA-MELANOGASTER CELL Raff, E. C., Fuller, M. T., Kaufman, T. C., Kemphues, K. J., RUDOLPH, J. E., Raff, R. A. 1982; 28 (1): 33-40

    Abstract

    Four different tubulins have been identified that are expressed during embryogenesis in Drosophila melanogaster. Two alpha-tubulin subunits (alpha 1 and alpha 2) and one beta-tubulin subunit (beta 1) are expressed throughout embryonic development. A second beta-tubulin subunit (beta 3) is expressed only for a short period in mid-embryonic development. Synthesis of beta 3-tubulin in vitro in a rabbit reticulocyte translation system is directed by RNA extracted from embryos only at the stage when the protein is expressed. Thus we conclude that the mRNA encoding beta 3-tubulin is transcribed only during the brief period of beta 3-tubulin synthesis. The expression of beta 3-tubulin is accompanied by a coordinate transient increase in the level of synthesis of the embryonic alpha-tubulins, thereby maintaining an approximately equimolar synthesis of alpha- and beta-tubulin subunits throughout embryogenesis.

    View details for Web of Science ID A1982MY42100007

    View details for PubMedID 6802501

  • PURIFICATION OF THE COAT AND SCAFFOLDING PROTEINS FROM PROCAPSIDS OF BACTERIOPHAGE P22 VIROLOGY Fuller, M. T., KING, J. 1981; 112 (2): 529-547

    View details for Web of Science ID A1981MA19200017

    View details for PubMedID 7257185

  • Investigations of bacteriophage P22 by laser Raman spectroscopy. Progress in clinical and biological research Li, Y., Thomas, G. J., Fuller, M., KING, J. 1981; 64: 271-283

    View details for PubMedID 7330048

  • Scaffolding proteins and the genetic control of virus shell assembly. Quarterly review of biology KING, J., Griffin-Shea, R., Fuller, M. T. 1980; 55 (4): 369-393

    Abstract

    Historically a gap has existed between the study of the one-dimensional organization of hereditary information in genes, and of the three-dimensional organization of macromolecules in biological structures. In this article we describe progress in closing this gap through the genetic and biochemical analysis of the assembly of the icosahedral shells of spherical viruses, a class of subcellular structures whose subunit organization is relatively well understood. The genes specifying the proteins required for capsid assembly have been identified for many bacterial viruses. By using mutants defective in these genes, it has been possible to identify intermediates in shell morphogenesis and DNA condensation, and to unravel the different levels of the genetic control of macromolecular assembly processes. In general, a precursor shell or procapsid is first constructed, and the DNA is subsequently coiled within it. The construction of a closed shell poses as difficult a problem for a virus as for an architect. In the well-studied bacteriophage P22 of Salmonella typhimurium, the construction of the procapsid requires the interaction of about 200 molecules of the gene-8 scaffolding protein with 420 molecules of the gene-5 coat protein, forming a double-shelled structure with the scaffolding protein on the inside. Once completed, procapsids undergo substantial alteration in the course of encapsulating the viral DNA. In P22, the initiation of DNA packaging triggers the exit of all of the scaffolding molecules from within the capsid, probably through the coat-protein lattice. These released molecules are re-utilized, interacting with newly synthesized coat subunits to form further procapsids. Thus, the scaffolding protein functions catalytically in capsid assembly. All of the well-studied DNA phages require a scaffolding protein species for procapsid assembly, though their properties vary. Purified coat and scaffolding subunits by themselves show little tendency to polymerize, and are stable as monomers in solution. Upon mixing together under the appropriate conditions, however, the proteins copolymerize into double shells. Their interaction with each other appears to be critical for efficient assembly; this interaction probably occurs on the edges of growing shells, and not among subunits in solution. We have termed this kind of process, which we previously described in T4 tail morphogenesis, self-regulated assembly. The subunits are synthesized in a nonreactive form and are activated, not in solution, but upon incorporation into the growing substrate structure. A number of further transformations of the capsid subunits occur only within the organized structure and not as free subunits. Thus, aspects of the genetic information controlling the assembly process are not fully expressed at the level of the properties of protein subunits, but become manifest only through interactions with other proteins, or at a higher level, after completion of the correct organized structure.

    View details for PubMedID 7267974

  • STUDIES OF VIRUS STRUCTURE BY LASER-RAMAN SPECTROSCOPY .5. INVESTIGATION OF SECONDARY STRUCTURES AND MACROMOLECULAR INTERACTIONS IN BACTERIOPHAGE-P22 BY LASER RAMAN-SPECTROSCOPY BIOPHYSICAL JOURNAL FISH, S. R., Hartman, K. A., Fuller, M. T., KING, J., THOMAS, G. J. 1980; 32 (1): 234-237

    Abstract

    Laser Raman spectra of the DNA bacteriophage P22 and of its precursor particles and related structures have been obtained using 514.5-nm excitation. The spectra show that P22 DNA exists in the B form both inside of the phage head and after extraction from the phage. The major coat protein (gp5) contains a secondary structure composed of 18% alpha-helix, 20% beta-sheet and 62% irregular conformations. The scaffolding protein (gp8) in the phage prohead is substantially richer than gp5 in alpha-helical content. Among the amino acid residues which give prominent Raman lines, the spectra show that tryptophans are exposed to solvent and most tyrosines are hydrogen bonded to positive donor groups. The above features of phage DNA and protein structures are nearly invariant to changes in temperature up to 80 degrees C, indicating a remarkable thermal stability of the phage head and its encapsulated DNA.

    View details for Web of Science ID A1980KM03600035

  • REGULATION OF COAT PROTEIN POLYMERIZATION BY THE SCAFFOLDING PROTEIN OF BACTERIOPHAGE-P22 BIOPHYSICAL JOURNAL Fuller, M. T., KING, J. 1980; 32 (1): 381-401

    Abstract

    In the morphogenesis of double stranded DNA phages, a precursor protein shell empty of DNA is first assembled and then filled with DNA. The assembly of the correctly dimensioned precursor shell (procapsid) of Salmonella bacteriophage P22 requires the interaction of some 420 coat protein subunits with approximately 200 scaffolding protein subunits to form a double shelled particle with the scaffolding protein on the inside. In the course of DNA packaging, all of the scaffolding protein subunits exit from the procapsid and participate in further rounds of procapsid assembly (King and Casjens. 1974. Nature (Lond.). 251:112-119). To study the mechanism of shell assembly we have purified the coat and scaffolding protein subunits by selective dissociation of isolated procapsids. Both proteins can be obtained as soluble subunits in Tris buffer at near neutral pH. The coat protein sedimented in sucrose gradients as a roughly spherical monomer, while the scaffolding protein sedimented as if it were an elongated monomer. When the two proteins were mixed together in 1.5 M guanidine hydrochloride and dialyzed back to buffer at room temperature, procapsids formed which were very similar in morphology, sedimentation behavior, and protein composition to procapsids formed in vivo. Incubation of either protein alone under the same conditions did not yield any large structures. We interpret these results to mean that the assembly of the shell involves a switching of both proteins from their nonaggregating to their aggregating forms through their mutual interaction. The results are discussed in terms of the general problem of self-regulated assembly and the control of protein polymerization in morphogenesis.

    View details for Web of Science ID A1980KM03600053

    View details for PubMedID 7018607

  • GENETIC-CONTROL OF ORGANELLE ASSEMBLY AT THE MOLECULAR-LEVEL .4. SCAFFOLDING PROTEINS AND THE GENETIC-CONTROL OF VIRUS SHELL ASSEMBLY QUARTERLY REVIEW OF BIOLOGY KING, J., GRIFFINSHEA, R., Fuller, M. T. 1980; 55 (4): 369-393