Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
1.
Genes Dev ; 38(13-14): 655-674, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39111825

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.


Subject(s)
Cell Lineage , Polyadenylation , Spermatocytes , Spermatogenesis , Animals , Polyadenylation/genetics , Male , Spermatogenesis/genetics , Spermatocytes/metabolism , Spermatocytes/cytology , Cell Lineage/genetics , Gene Expression Regulation, Developmental/genetics , Adult Stem Cells/metabolism , Adult Stem Cells/cytology , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Spermatogonia/cytology , Spermatogonia/metabolism , mRNA Cleavage and Polyadenylation Factors/metabolism , mRNA Cleavage and Polyadenylation Factors/genetics
2.
Genes Dev ; 36(15-16): 916-935, 2022 08 01.
Article in English | MEDLINE | ID: mdl-36175033

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.


Subject(s)
Adult Stem Cells , RNA Isoforms , 3' Untranslated Regions/genetics , Adult Stem Cells/metabolism , Animals , Male , Polyadenylation , Protein Isoforms/genetics , RNA Isoforms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
3.
Genes Dev ; 34(9-10): 663-677, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32217666

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 ß2UE1 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.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/growth & development , Drosophila melanogaster/genetics , Spermatogenesis/genetics , Amino Acid Motifs/genetics , Animals , Base Sequence/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Male , Promoter Regions, Genetic/genetics , Spermatocytes/cytology , Spermatocytes/metabolism , Transcription Initiation Site , Transcriptome/genetics
4.
Proc Natl Acad Sci U S A ; 121(42): e2309548121, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39378093

ABSTRACT

Posttranscriptional regulation of gene expression by RNA-binding proteins can enhance the speed and robustness of cell state transitions by controlling RNA stability, localization, or if, when, or where mRNAs are translated. The RNA helicase YTHDC2 is required to shut down components of the mitotic program to facilitate a proper switch from mitosis to meiosis in mouse germ cells. Here, we show that YTHDC2 has a second essential role in promoting meiotic progression in late spermatocytes. Inducing conditional knockout of Ythdc2 during the first wave of spermatogenesis, after initiation of meiotic prophase, allowed YTHDC2-deficient germ cells to advance to the pachytene stage and properly express many meiotic markers. However, the YTHDC2-deficient spermatocytes mis-expressed a number of genes, some up-regulated and some down-regulated, failed to transition to the diplotene stage, and then quickly died. Coimmunoprecipitation experiments revealed that YTHDC2 interacts with several RNA-binding proteins in early or late spermatocytes, with many of the interacting proteins, including MEIOC, localizing to granules, similar to YTHDC2. Our findings suggest that YTHDC2 collaborates with other RNA granule components to facilitate proper progression of germ cells through multiple steps of meiosis via mechanisms influencing posttranscriptional regulation of RNAs.


Subject(s)
Meiosis , RNA Helicases , RNA-Binding Proteins , Spermatocytes , Spermatogenesis , Animals , Male , Spermatocytes/metabolism , Spermatocytes/cytology , Mice , Spermatogenesis/physiology , Spermatogenesis/genetics , Meiosis/physiology , RNA Helicases/metabolism , RNA Helicases/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Cell Differentiation , Mice, Knockout , Germ Cells/metabolism
5.
Development ; 150(22)2023 Nov 15.
Article in English | MEDLINE | ID: mdl-37882771

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 over 1800 genes and grow 25-fold. Previous work has shown 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-h window just before spermatocytes are fully mature. In males mutant for rbp4 or lut, spermatocytes enter and exit meiotic division 6-8 h 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.


Subject(s)
Drosophila Proteins , Meiosis , Animals , Male , Meiosis/genetics , Spermatogenesis/physiology , Prophase , Mitosis , Spermatocytes/metabolism , Drosophila/genetics , Cyclin B/genetics , Cyclin B/metabolism , Drosophila Proteins/metabolism
6.
RNA Biol ; 20(1): 908-925, 2023 01.
Article in English | MEDLINE | ID: mdl-37906624

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.


Subject(s)
Gene Expression Regulation , Polyadenylation , Humans , 3' Untranslated Regions , RNA Isoforms/genetics , RNA Isoforms/metabolism , Cell Differentiation/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
8.
Development ; 146(8)2019 04 15.
Article in English | MEDLINE | ID: mdl-30918053

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 the 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 the function of the Par complex had been knocked down survived and proliferated, 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 the 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.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Drosophila Proteins/metabolism , Drosophila/cytology , Drosophila/metabolism , Germ Cells/cytology , Germ Cells/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Protein Kinase C/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Polarity/genetics , Cell Polarity/physiology , Drosophila Proteins/genetics , Intracellular Signaling Peptides and Proteins/genetics , Male , Mitosis/genetics , Mitosis/physiology , Protein Kinase C/genetics
9.
Development ; 146(22)2019 11 18.
Article in English | MEDLINE | ID: mdl-31645358

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 of 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.


Subject(s)
Drosophila Proteins/physiology , Drosophila melanogaster/embryology , Gene Expression Regulation, Developmental , Intracellular Signaling Peptides and Proteins/physiology , Meiosis , RNA, Messenger/genetics , Spermatogenesis , Animals , Axoneme/metabolism , Cell Nucleus/metabolism , Centrosome/metabolism , Chromosome Segregation , Cloning, Molecular , Crosses, Genetic , Cyclin B , Cytokinesis , Drosophila Proteins/genetics , In Situ Hybridization, Fluorescence , Intracellular Signaling Peptides and Proteins/genetics , Male , Microtubules/metabolism , Mutation , RNA-Binding Proteins , Spermatocytes/metabolism , Spindle Apparatus/metabolism , Transgenes , Zinc Fingers
10.
PLoS Genet ; 15(6): e1008187, 2019 06.
Article in English | MEDLINE | ID: mdl-31226128

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.


Subject(s)
Adenosine Triphosphatases/genetics , Adult Stem Cells/metabolism , Autoantigens/genetics , Drosophila Proteins/genetics , Retinoblastoma-Binding Protein 4/genetics , Transcription Factors/genetics , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Cell Self Renewal/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Germ Cells/growth & development , Male , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Stem Cell Niche/genetics , Testis/growth & development , Testis/metabolism , Transforming Growth Factor beta/genetics
11.
Development ; 142(19): 3394-402, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26443637

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.


Subject(s)
Cyclin B/metabolism , Drosophila/embryology , Gene Expression Regulation, Developmental/physiology , Meiosis/physiology , Spermatogenesis/physiology , Animals , Animals, Genetically Modified , Blotting, Western , CRISPR-Cas Systems , Cloning, Molecular , DNA Primers/genetics , Drosophila/genetics , Drosophila Proteins/metabolism , Immunoprecipitation , Male , Microscopy, Fluorescence , RNA Polymerase II/metabolism , Reverse Transcriptase Polymerase Chain Reaction
12.
PLoS Genet ; 11(12): e1005701, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26624996

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.


Subject(s)
Drosophila Proteins/genetics , Mediator Complex/genetics , Spermatogenesis , Transcription Factor TFIID/genetics , Transcription Factors/genetics , Transcriptional Activation/genetics , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Animals , Cell Differentiation/genetics , Cell Lineage , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Gene Expression Regulation, Developmental , Germ Cells/cytology , Germ Cells/metabolism , Male , Mediator Complex/metabolism , Meiosis/genetics , Spermatogonia/growth & development , Testis/growth & development
13.
PLoS Genet ; 11(11): e1005632, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26528720

ABSTRACT

Mitotic and cytokinetic processes harness cell machinery to drive chromosomal segregation and the physical separation of dividing cells. Here, we investigate the functional requirements for exocyst complex function during cell division in vivo, and demonstrate a common mechanism that directs anaphase cell elongation and cleavage furrow progression during cell division. We show that onion rings (onr) and funnel cakes (fun) encode the Drosophila homologs of the Exo84 and Sec8 exocyst subunits, respectively. In onr and fun mutant cells, contractile ring proteins are recruited to the equatorial region of dividing spermatocytes. However, cytokinesis is disrupted early in furrow ingression, leading to cytokinesis failure. We use high temporal and spatial resolution confocal imaging with automated computational analysis to quantitatively compare wild-type versus onr and fun mutant cells. These results demonstrate that anaphase cell elongation is grossly disrupted in cells that are compromised in exocyst complex function. Additionally, we observe that the increase in cell surface area in wild type peaks a few minutes into cytokinesis, and that onr and fun mutant cells have a greatly reduced rate of surface area growth specifically during cell division. Analysis by transmission electron microscopy reveals a massive build-up of cytoplasmic astral membrane and loss of normal Golgi architecture in onr and fun spermatocytes, suggesting that exocyst complex is required for proper vesicular trafficking through these compartments. Moreover, recruitment of the small GTPase Rab11 and the PITP Giotto to the cleavage site depends on wild-type function of the exocyst subunits Exo84 and Sec8. Finally, we show that the exocyst subunit Sec5 coimmunoprecipitates with Rab11. Our results are consistent with the exocyst complex mediating an essential, coordinated increase in cell surface area that potentiates anaphase cell elongation and cleavage furrow ingression.


Subject(s)
Anaphase , Cell Cycle , Drosophila/cytology , Animals
14.
BMC Cell Biol ; 18(1): 16, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28335714

ABSTRACT

BACKGROUND: 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. RESULTS: 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. CONCLUSIONS: 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.


Subject(s)
Drosophila Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proton-Translocating ATPases/metabolism , Morphogenesis , Protein Subunits/metabolism , Proton-Translocating ATPases/metabolism , Testis/embryology , Animals , Drosophila melanogaster/enzymology , Evolution, Molecular , Flight, Animal/physiology , Gene Knockdown Techniques , Genes, Insect , Green Fluorescent Proteins/metabolism , Male , Mitochondrial Proton-Translocating ATPases/chemistry , Mitochondrial Proton-Translocating ATPases/genetics , Models, Biological , Muscle, Skeletal/metabolism , Mutation/genetics , Organ Specificity , Phenotype , Phylogeny , Protein Multimerization , Protein Subunits/genetics , RNA Interference , Spermatids/metabolism , Spermatogenesis
15.
Development ; 141(1): 73-82, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24346697

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.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Profilins/metabolism , Spermatozoa/metabolism , Stem Cells/metabolism , Tumor Suppressor Proteins/metabolism , Actins/metabolism , Adherens Junctions/metabolism , Animals , Cadherins/metabolism , Cell Differentiation , Cell Lineage , Cell Proliferation , Drosophila Proteins/biosynthesis , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Gene Expression Regulation, Developmental , Male , Profilins/genetics , Protein Binding , RNA Interference , RNA, Small Interfering/genetics , Signal Transduction/genetics , Spermatogonia/cytology , Spermatogonia/metabolism , Spermatozoa/cytology , Stem Cell Niche , Tumor Suppressor Proteins/biosynthesis
16.
PLoS Genet ; 10(5): e1004305, 2014 May.
Article in English | MEDLINE | ID: mdl-24786584

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.


Subject(s)
Cytokinesis , Drosophila Proteins/physiology , Drosophila melanogaster/cytology , rab GTP-Binding Proteins/metabolism , Amino Acid Sequence , Animals , Binding Sites , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Humans , Molecular Sequence Data , Mutation , Sequence Homology, Amino Acid , Spindle Apparatus
17.
Development ; 140(17): 3522-31, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23884444

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.


Subject(s)
Cell Cycle/physiology , Cell Differentiation/physiology , Drosophila/growth & development , Germ Cells/growth & development , Meiosis/physiology , Polyubiquitin/genetics , Animals , Blotting, Western , Chromosome Mapping , Cloning, Molecular , DNA Primers/genetics , In Situ Hybridization , Male , Microarray Analysis , Microscopy, Fluorescence , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Testis/growth & development
18.
PLoS Genet ; 9(11): e1003903, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24244183

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.


Subject(s)
Adult Stem Cells/metabolism , Chromatin Assembly and Disassembly/genetics , Drosophila Proteins/metabolism , Histones/genetics , Transcription Factors/metabolism , Adult Stem Cells/cytology , Animals , Cell Differentiation , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Germ Cells , Homeostasis , Male , Signal Transduction , Testis/metabolism , Transcription Factors/genetics
19.
Development ; 139(8): 1381-90, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22378638

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.


Subject(s)
Germ Cells/cytology , Stem Cells/cytology , Adherens Junctions/metabolism , Animals , Cadherins/metabolism , Cell Adhesion , Cell Differentiation , Cell Lineage , Crosses, Genetic , Drosophila Proteins/metabolism , Drosophila melanogaster , Female , Green Fluorescent Proteins/metabolism , Male , Microscopy, Phase-Contrast/methods , Receptor-Like Protein Tyrosine Phosphatases/metabolism
20.
Proc Natl Acad Sci U S A ; 109(45): 18477-81, 2012 Nov 06.
Article in English | MEDLINE | ID: mdl-23091022

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.


Subject(s)
Cell Differentiation , Cell Lineage , Drosophila melanogaster/cytology , Germ Cells/cytology , Stem Cells/cytology , Testis/cytology , Animals , Drosophila melanogaster/metabolism , Germ Cells/metabolism , Male , Signal Transduction , Stem Cells/metabolism , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL