Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Nucleic Acids Res ; 47(22): 11649-11666, 2019 12 16.
Article in English | MEDLINE | ID: mdl-31701127

ABSTRACT

CoREST has been identified as a subunit of several protein complexes that generate transcriptionally repressive chromatin structures during development. However, a comprehensive analysis of the CoREST interactome has not been carried out. We use proteomic approaches to define the interactomes of two dCoREST isoforms, dCoREST-L and dCoREST-M, in Drosophila. We identify three distinct histone deacetylase complexes built around a common dCoREST/dRPD3 core: A dLSD1/dCoREST complex, the LINT complex and a dG9a/dCoREST complex. The latter two complexes can incorporate both dCoREST isoforms. By contrast, the dLSD1/dCoREST complex exclusively assembles with the dCoREST-L isoform. Genome-wide studies show that the three dCoREST complexes associate with chromatin predominantly at promoters. Transcriptome analyses in S2 cells and testes reveal that different cell lineages utilize distinct dCoREST complexes to maintain cell-type-specific gene expression programmes: In macrophage-like S2 cells, LINT represses germ line-related genes whereas other dCoREST complexes are largely dispensable. By contrast, in testes, the dLSD1/dCoREST complex prevents transcription of germ line-inappropriate genes and is essential for spermatogenesis and fertility, whereas depletion of other dCoREST complexes has no effect. Our study uncovers three distinct dCoREST complexes that function in a lineage-restricted fashion to repress specific sets of genes thereby maintaining cell-type-specific gene expression programmes.


Subject(s)
Chromatin/metabolism , Co-Repressor Proteins/metabolism , Drosophila Proteins/metabolism , Gene Expression Regulation/genetics , Histone Deacetylases/metabolism , Oxidoreductases, N-Demethylating/metabolism , Animals , Cell Line , Drosophila melanogaster/embryology , Epigenesis, Genetic/genetics , Gene Expression Profiling , Histone Deacetylases/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Protein Isoforms/genetics , Transcription Factors/metabolism , Transcriptome/genetics
2.
Development ; 143(2): 329-38, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26657767

ABSTRACT

The testis of Drosophila resembles an individual testis tubule of mammals. Both are surrounded by a sheath of smooth muscles, which in Drosophila are multinuclear and originate from a pool of myoblasts that are set aside in the embryo and accumulate on the genital disc later in development. These muscle stem cells start to differentiate early during metamorphosis and give rise to all muscles of the inner male reproductive system. Shortly before the genital disc and the developing testes connect, multinuclear nascent myotubes appear on the anterior tips of the seminal vesicles. Here, we show that adhesion molecules are distinctly localized on the seminal vesicles; founder cell (FC)-like myoblasts express Dumbfounded (Duf) and Roughest (Rst), and fusion-competent myoblast (FCM)-like cells mainly express Sticks and stones (Sns). The smooth but multinuclear myotubes of the testes arose by myoblast fusion. RNAi-mediated attenuation of Sns or both Duf and Rst severely reduced the number of nuclei in the testes muscles. Duf and Rst probably act independently in this context. Despite reduced fusion in all of these RNAi-treated animals, myotubes migrated onto the testes, testes were shaped and coiled, muscle filaments were arranged as in the wild type and spermatogenesis proceeded normally. Hence, the testes muscles compensate for fusion defects so that the myofibres encircling the adult testes are indistinguishable from those of the wild type and male fertility is guaranteed.


Subject(s)
Drosophila Proteins/metabolism , Myoblasts/cytology , Testis/cytology , Animals , Drosophila , Drosophila Proteins/genetics , Male , Models, Biological , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Myoblasts/physiology , Testis/physiology
3.
Nucleic Acids Res ; 43(6): 3033-45, 2015 Mar 31.
Article in English | MEDLINE | ID: mdl-25735749

ABSTRACT

Despite insights on the cellular level, the molecular details of chromatin reorganization in sperm development, which involves replacement of histone proteins by specialized factors to allow ultra most condensation of the genome, are not well understood. Protamines are dispensable for DNA condensation during Drosophila post-meiotic spermatogenesis. Therefore, we analyzed the interaction of Mst77F, another very basic testis-specific protein with chromatin and DNA as well as studied the molecular consequences of such binding. We show that Mst77F on its own causes severe chromatin and DNA aggregation. An intrinsically unstructured domain in the C-terminus of Mst77F binds DNA via electrostatic interaction. This binding results in structural reorganization of the domain, which induces interaction with an N-terminal region of the protein. Via putative cooperative effects Mst77F is induced to multimerize in this state causing DNA aggregation. In agreement, overexpression of Mst77F results in chromatin aggregation in fly sperm. Based on these findings we postulate that Mst77F is crucial for sperm development by giving rise to a unique condensed chromatin structure.


Subject(s)
Chromatin/chemistry , Chromatin/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Histones/chemistry , Histones/metabolism , Animals , Animals, Genetically Modified , Chromatin/genetics , Chromatin Assembly and Disassembly , DNA/chemistry , DNA/genetics , DNA/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Histones/genetics , Male , Mutagenesis, Site-Directed , Protamines/metabolism , Protein Interaction Domains and Motifs , Protein Multimerization , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Spermatozoa/metabolism , Static Electricity
4.
Biochim Biophys Acta ; 1839(3): 155-68, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24091090

ABSTRACT

The function of sperm is to safely transport the haploid paternal genome to the egg containing the maternal genome. The subsequent fertilization leads to transmission of a new unique diploid genome to the next generation. Before the sperm can set out on its adventurous journey, remarkable arrangements need to be made during the post-meiotic stages of spermatogenesis. Haploid spermatids undergo extensive morphological changes, including a striking reorganization and compaction of their chromatin. Thereby, the nucleosomal, histone-based structure is nearly completely substituted by a protamine-based structure. This replacement is likely facilitated by incorporation of histone variants, post-translational histone modifications, chromatin-remodeling complexes, as well as transient DNA strand breaks. The consequences of mutations have revealed that a protamine-based chromatin is essential for fertility in mice but not in Drosophila. Nevertheless, loss of protamines in Drosophila increases the sensitivity to X-rays and thus supports the hypothesis that protamines are necessary to protect the paternal genome. Pharmaceutical approaches have provided the first mechanistic insights and have shown that hyperacetylation of histones just before their displacement is vital for progress in chromatin reorganization but is clearly not the sole inducer. In this review, we highlight the current knowledge on post-meiotic chromatin reorganization and reveal for the first time intriguing parallels in this process in Drosophila and mammals. We conclude with a model that illustrates the possible mechanisms that lead from a histone-based chromatin to a mainly protamine-based structure during spermatid differentiation. This article is part of a Special Issue entitled: Chromatin and epigenetic regulation of animal development.


Subject(s)
Chromatin Assembly and Disassembly/physiology , Genomic Instability/physiology , Protein Processing, Post-Translational/physiology , Spermatids/metabolism , Spermatogenesis/physiology , Animals , DNA Breaks , Drosophila melanogaster , Histones/metabolism , Humans , Male , Mice , Nucleosomes/metabolism , Spermatids/cytology
5.
Dev Biol ; 377(1): 33-45, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23466740

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.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Gene Expression Regulation , Histones/genetics , Protamines/genetics , 5' Untranslated Regions/genetics , Animals , Base Sequence , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Histones/metabolism , Male , Open Reading Frames/genetics , Protamines/metabolism , Protein Biosynthesis/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spermatocytes/cytology , Spermatocytes/metabolism , TATA-Binding Protein Associated Factors/metabolism , Transcription, Genetic
6.
BMC Cell Biol ; 15: 27, 2014 Jul 08.
Article in English | MEDLINE | ID: mdl-25000973

ABSTRACT

BACKGROUND: The visceral musculature of Drosophila larvae comprises circular visceral muscles tightly interwoven with longitudinal visceral muscles. During myogenesis, the circular muscles arise by one-to-one fusion of a circular visceral founder cell (FC) with a visceral fusion-competent myoblast (FCM) from the trunk visceral mesoderm, and longitudinal muscles arise from FCs of the caudal visceral mesoderm. Longitudinal FCs migrate anteriorly under guidance of fibroblast growth factors during embryogenesis; it is proposed that they fuse with FCMs from the trunk visceral mesoderm to give rise to syncytia containing up to six nuclei. RESULTS: Using fluorescence in situ hybridization and immunochemical analyses, we investigated whether these fusion events during migration use the same molecular repertoire and cellular components as fusion-restricted myogenic adhesive structure (FuRMAS), the adhesive signaling center that mediates myoblast fusion in the somatic mesoderm. Longitudinal muscles were formed by the fusion of one FC with Sns-positive FCMs, and defects in FCM specification led to defects in longitudinal muscle formation. At the fusion sites, Duf/Kirre and the adaptor protein Rols7 accumulated in longitudinal FCs, and Blow and F-actin accumulated in FCMs. The accumulation of these four proteins at the fusion sites argues for FuRMAS-like adhesion and signaling centers. Longitudinal fusion was disturbed in rols and blow single, and scar wip double mutants. Mutants of wasp or its interaction partner wip had no defects in longitudinal fusion. CONCLUSIONS: Our results indicated that all embryonic fusion events depend on the same cell-adhesion molecules, but that the need for Rols7 and regulators of F-actin distinctly differs. Rols7 was required for longitudinal visceral and somatic myoblast fusion but not for circular visceral fusion. Importantly, longitudinal fusion depended on Kette and SCAR/Wave but was independent of WASp-dependent Arp2/3 activation. Thus, the complexity of the players involved in muscle formation increases from binucleated circular muscles to longitudinal visceral muscles to somatic muscles.


Subject(s)
Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Myoblasts/cytology , Animals , Animals, Genetically Modified , Cell Movement , Drosophila Proteins/analysis , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Gene Expression Regulation, Developmental , In Situ Hybridization, Fluorescence , Muscle Development , Muscle Proteins/analysis , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscles/embryology , Muscles/metabolism , Myoblasts/metabolism
7.
J Cell Sci ; 125(Pt 23): 5667-76, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22976306

ABSTRACT

Injury is an inevitable part of life, making wound healing essential for survival. In postembryonic skin, wound closure requires that epidermal cells recognize the presence of a gap and change their behavior to migrate across it. In Drosophila larvae, wound closure requires two signaling pathways [the Jun N-terminal kinase (JNK) pathway and the Pvr receptor tyrosine kinase signaling pathway] and regulation of the actin cytoskeleton. In this and other systems, it remains unclear how the signaling pathways that initiate wound closure connect to the actin regulators that help execute wound-induced cell migrations. Here, we show that chickadee, which encodes the Drosophila Profilin, a protein important for actin filament recycling and cell migration during development, is required for the physiological process of larval epidermal wound closure. After injury, chickadee is transcriptionally upregulated in cells proximal to the wound. We found that JNK, but not Pvr, mediates the increase in chic transcription through the Jun and Fos transcription factors. Finally, we show that chic-deficient larvae fail to form a robust actin cable along the wound edge and also fail to form normal filopodial and lamellipodial extensions into the wound gap. Our results thus connect a factor that regulates actin monomer recycling to the JNK signaling pathway during wound closure. They also reveal a physiological function for an important developmental regulator of actin and begin to tease out the logic of how the wound repair response is organized.


Subject(s)
Larva/genetics , Profilins/genetics , Wound Healing/physiology , Animals , Animals, Genetically Modified , Cell Movement/genetics , Cell Movement/physiology , Drosophila , Drosophila Proteins/genetics , Wound Healing/genetics
8.
J Neurogenet ; 28(3-4): 302-15, 2014.
Article in English | MEDLINE | ID: mdl-24957080

ABSTRACT

Membrane fusion is essential for the communication of membrane-defined compartments, development of multicellular organisms and tissue homeostasis. Although membrane fusion has been studied extensively, still little is known about the molecular mechanisms. Especially the intercellular fusion of cells during development and tissue homeostasis is poorly understood. Somatic muscle formation in Drosophila depends on the intercellular fusion of myoblasts. In this process, myoblasts recognize each other and adhere, thereby triggering a protein machinery that leads to electron-dense plaques, vesicles and F-actin formation at apposing membranes. Two models of how local membrane stress is achieved to induce the merging of the myoblast membranes have been proposed: the electron-dense vesicles transport and release a fusogen and F-actin bends the plasma membrane. In this review, we highlight cell-adhesion molecules and intracellular proteins known to be involved in myoblast fusion. The cell-adhesion proteins also mediate the recognition and adhesion of other cell types, such as neurons that communicate with each other via special intercellular junctions, termed chemical synapses. At these synapses, neurotransmitters are released through the intracellular fusion of synaptic vesicles with the plasma membrane. As the targeting of electron-dense vesicles in myoblasts shares some similarities with the targeting of synaptic vesicle fusion, we compare molecules required for synaptic vesicle fusion to recently identified molecules involved in myoblast fusion.


Subject(s)
Cell Membrane/metabolism , Myoblasts/metabolism , Synapses/metabolism , Animals , Cell Adhesion Molecules/metabolism , Drosophila/metabolism , Exocytosis/physiology , Synaptic Vesicles/metabolism
9.
Exp Cell Res ; 319(4): 402-16, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23246571

ABSTRACT

Besides representing the sarcomeric thick filaments, myosins are involved in many cellular transport and motility processes. Myosin heavy chains are grouped into 18 classes. Here we show that in Drosophila, the unconventional group XVIII myosin heavy chain-like (Mhcl) is transcribed in the mesoderm of embryos, most prominently in founder cells (FCs). An ectopically expressed GFP-tagged Mhcl localizes in the growing muscle at cell-cell contacts towards the attached fusion competent myoblast (FCM). We further show that Mhcl interacts in vitro with the essential fusion protein Rolling pebbles 7 (Rols7), which is part of a protein complex established at cell contact sites (Fusion-restricted Myogenic-Adhesive Structure or FuRMAS). Here, branched F-actin is likely needed to widen the fusion pore and to integrate the myoblast into the growing muscle. We show that the localization of Mhcl is dependent on the presence of Rols7, and we postulate that Mhcl acts at the FuRMAS as an actin motor protein. We further show that Mhcl deficient embryos develop a wild-type musculature. We thus propose that Mhcl functions redundantly to other myosin heavy chains in myoblasts. Lastly, we found that the protein is detectable adjacent to the sarcomeric Z-discs, suggesting an additional function in mature muscles.


Subject(s)
Cell Communication , Drosophila Proteins/metabolism , Drosophila melanogaster , Membrane Proteins/metabolism , Muscle Proteins/metabolism , Myoblasts/physiology , Myosins/metabolism , Animals , Animals, Genetically Modified , Cell Adhesion/genetics , Cell Communication/genetics , Cell Communication/physiology , Cell Fusion , Cells, Cultured , Drosophila Proteins/genetics , Drosophila Proteins/physiology , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Embryo, Nonmammalian , Gene Expression Regulation, Developmental , Membrane Proteins/genetics , Membrane Proteins/physiology , Muscle Development/genetics , Muscle Development/physiology , Muscle Proteins/genetics , Muscle Proteins/physiology , Myoblasts/metabolism , Myosins/genetics , Protein Binding/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/physiology , Protein Transport , Tissue Distribution
10.
PLoS Genet ; 7(7): e1002206, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21829383

ABSTRACT

Eukaryotic cells respond to genomic and environmental stresses, such as DNA damage and heat shock (HS), with the synthesis of poly-[ADP-ribose] (PAR) at specific chromatin regions, such as DNA breaks or HS genes, by PAR polymerases (PARP). Little is known about the role of this modification during cellular stress responses. We show here that the nucleosome remodeler dMi-2 is recruited to active HS genes in a PARP-dependent manner. dMi-2 binds PAR suggesting that this physical interaction is important for recruitment. Indeed, a dMi-2 mutant unable to bind PAR does not localise to active HS loci in vivo. We have identified several dMi-2 regions which bind PAR independently in vitro, including the chromodomains and regions near the N-terminus containing motifs rich in K and R residues. Moreover, upon HS gene activation, dMi-2 associates with nascent HS gene transcripts, and its catalytic activity is required for efficient transcription and co-transcriptional RNA processing. RNA and PAR compete for dMi-2 binding in vitro, suggesting a two step process for dMi-2 association with active HS genes: initial recruitment to the locus via PAR interaction, followed by binding to nascent RNA transcripts. We suggest that stress-induced chromatin PARylation serves to rapidly attract factors that are required for an efficient and timely transcriptional response.


Subject(s)
Adenosine Triphosphatases/metabolism , Autoantigens/metabolism , Drosophila Proteins/metabolism , Drosophila/genetics , Drosophila/metabolism , Gene Expression Regulation , Heat-Shock Response/genetics , Poly(ADP-ribose) Polymerases/metabolism , Stress, Physiological , Adenosine Triphosphatases/genetics , Amino Acid Sequence , Animals , Autoantigens/genetics , Drosophila Proteins/genetics , Models, Biological , Molecular Sequence Data , Poly Adenosine Diphosphate Ribose/metabolism , Protein Binding/physiology , Protein Interaction Domains and Motifs , RNA/genetics , RNA/metabolism , Sequence Alignment , Transcription, Genetic
11.
Dev Biol ; 370(1): 86-97, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22841645

ABSTRACT

The adult musculature in D. melanogaster forms during metamorphosis. Much is known about the flight and leg musculature, but not about the muscles surrounding the male reproductive tract. The inner genitalia of males consist of the testes, which emerge from the gonads; the remaining genital organs, i.e., paragonia (or accessory glands), ejaculatory duct, sperm pump, and seminal vesicles, develop out of the genital imaginal disc. We analyzed the myoblasts forming the muscle layers of these organs. In myoblasts derived from the genital imaginal disc, the regulatory region of the transcription factor DMef2 is active. DMef2 is also needed for specification and differentiation of embryonic and adult myoblasts. We could discriminate three different muscle types: (i) multinucleated muscles that resemble vertebrate smooth muscles surround the testes, (ii) multinucleated muscles that resemble striated muscles comprises seminal vesicles and the sperm pump, and (iii) mononucleated striated musculature encloses the paragonia and ejaculatory duct. Members of the immunoglobulin superfamily involved in embryonic myogenesis, Dumbfounded (Duf) and Sticks and Stones (Sns), were also expressed in the genital imaginal disc, in the muscle sheath of the testes during muscle differentiation and in the secretory secondary cells, which are part of the binucleated epithelia enclosing the paragonia.


Subject(s)
Drosophila melanogaster/growth & development , Gene Expression Regulation, Developmental/physiology , Genitalia, Male/growth & development , Metamorphosis, Biological/physiology , Muscle Development/physiology , Muscle, Smooth/growth & development , Muscle, Striated/growth & development , Animals , Cell Differentiation/physiology , Drosophila Proteins/metabolism , Gene Expression Regulation, Developmental/genetics , Genitalia, Male/ultrastructure , Immunoglobulins/metabolism , Immunohistochemistry , Male , Membrane Proteins/metabolism , Microscopy, Electron, Scanning , Muscle Proteins/metabolism , Myoblasts/cytology , Myoblasts/ultrastructure
12.
Dev Genes Evol ; 223(3): 159-69, 2013 May.
Article in English | MEDLINE | ID: mdl-23111653

ABSTRACT

Fibroblast growth factor receptors (FGFR) are highly conserved receptor tyrosine kinases, and evolved early in metazoan evolution. In order to investigate their functional conservation, we asked whether the Kringelchen FGFR in the freshwater polyp Hydra vulgaris, is able to functionally replace FGFR in fly embryos. In Drosophila, two endogenous FGFR, Breathless (Btl) and Heartless (Htl), ensure formation of the tracheal system and mesodermal cell migration as well as formation of the heart. Using UAS-kringelchen-5xmyc transgenic flies and targeted expression, we show that Kringelchen is integrated correctly into the cell membrane of mesodermal and tracheal cells in Drosophila. Nevertheless, Kringelchen expression driven in tracheal cells failed to rescue the btl (LG19) mutant. The Hydra FGFR was able to substitute for Heartless in the htl (AB42) null mutant; however, this occurred only during early mesodermal cell migration. Our data provide evidence for functional conservation of this early-diverged FGFR across these distantly related phyla, but also selectivity for the Htl FGFR in the Drosophila system.


Subject(s)
Drosophila/genetics , Hydra/genetics , Receptors, Fibroblast Growth Factor/genetics , Amino Acid Sequence , Animals , Animals, Genetically Modified , Evolution, Molecular , Molecular Sequence Data , Mutation , Phylogeny , Receptors, Fibroblast Growth Factor/chemistry , Sequence Homology, Amino Acid
13.
J Cell Sci ; 124(Pt 19): 3266-78, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21896648

ABSTRACT

In the Drosophila embryo, transient cell adhesion during myoblast fusion is known to lead to the formation of fusion-restricted myogenic-adhesive structures (FuRMASs). Here, we report that within these FuRMASs, a Drosophila homologue of human and mouse swiprosins (EF-hand-domain-containing proteins) is expressed, which we named Drosophila Swiprosin-1 (Drosophila Swip-1). Drosophila Swip-1 is highly conserved and is closely related to the calcium-binding proteins swiprosin-1 and swiprosin-2 that have a role in the immune system in humans and mice. Our study shows that Drosophila Swip-1 is also expressed in corresponding cells of the Drosophila immune system. During myoblast fusion, Drosophila Swip-1 accumulates transiently in the foci of fusion-competent myoblasts (FCMs). Both the EF-hand and the coiled-coil domain of Drosophila Swip-1 are required to localise the protein to these foci. The formation of Drosophila Swip-1 foci requires successful cell adhesion between FCMs and founder cells (FCs) or growing myotubes. Moreover, Drosophila Swip-1 foci were found to increase in number in sing(22) mutants, which arrest myoblast fusion after prefusion complex formation. By contrast, Drosophila Swip-1 foci are not significantly enriched in blow(2) and kette(J4-48) mutants, which stop myogenesis beyond the prefusion complex stage but before plasma membrane merging. Therefore, we hypothesise that Drosophila Swip-1 participates in the breakdown of the prefusion complex during the progression of myoblast fusion.


Subject(s)
Drosophila melanogaster/cytology , Myoblasts/physiology , Amino Acid Sequence , Animals , Cell Adhesion , Cell Fusion , Cell Membrane/metabolism , Conserved Sequence , Cytoplasmic Vesicles/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , Exocytosis , Female , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Immune System/metabolism , Immunoglobulins/genetics , Immunoglobulins/metabolism , Inclusion Bodies/metabolism , Larva/cytology , Larva/immunology , Larva/metabolism , Mesoderm/metabolism , Molecular Sequence Data , Muscle Development , Myoblasts/metabolism , Protein Structure, Tertiary , Protein Transport , RNA Interference , Sequence Deletion , Signal Transduction
14.
Nat Commun ; 12(1): 791, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33542237

ABSTRACT

Cells migrate collectively to form tissues and organs during morphogenesis. Contact inhibition of locomotion (CIL) drives collective migration by inhibiting lamellipodial protrusions at cell-cell contacts and promoting polarization at the leading edge. Here, we report a CIL-related collective cell behavior of myotubes that lack lamellipodial protrusions, but instead use filopodia to move as a cohesive cluster in a formin-dependent manner. We perform genetic, pharmacological and mechanical perturbation analyses to reveal the essential roles of Rac2, Cdc42 and Rho1 in myotube migration. These factors differentially control protrusion dynamics and cell-matrix adhesion formation. We also show that active Rho1 GTPase localizes at retracting free edge filopodia and that Rok-dependent actomyosin contractility does not mediate a contraction of protrusions at cell-cell contacts, but likely plays an important role in the constriction of supracellular actin cables. Based on these findings, we propose that contact-dependent asymmetry of cell-matrix adhesion drives directional movement, whereas contractile actin cables contribute to the integrity of the migrating cell cluster.


Subject(s)
Cell Movement/physiology , Morphogenesis/physiology , Muscle Fibers, Skeletal/physiology , Pseudopodia/metabolism , Actin Cytoskeleton/metabolism , Actomyosin/metabolism , Animals , Cadherins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster , GTP-Binding Proteins/metabolism , Intravital Microscopy , rac GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/metabolism , RAC2 GTP-Binding Protein
15.
PLoS One ; 14(3): e0213177, 2019.
Article in English | MEDLINE | ID: mdl-30845228

ABSTRACT

Spermatogenesis in Drosophila melanogaster is characterized by a specific transcriptional program during the spermatocyte stage. Transcription of thousands of genes is regulated by the interaction of several proteins or complexes, including a tTAF-containing TFIID variant, tMAC, Mediator, and chromatin interactors, e.g., bromodomain proteins. We addressed how distinct subsets of target genes are selected. We characterized the highly similar proteins tPlus3a and tPlus3b, which contain a Plus3 domain and are enriched in the testis, mainly in spermatocytes. In tPlus3a and tplus3b deletion mutants generated using the CRISPR/Cas9 system, fertility was severely reduced and sperm showed defects during individualization. tPlus3a and tPlus3b heterodimerized with the bromodomain protein tBRD-1. To elucidate the role of the tPlus3a and tPlus3b proteins in transcriptional regulation, we determined the transcriptomes of tplus3a-tplus3b and tbrd-1 deletion mutants using next-generation sequencing (RNA-seq) and compared them to that of the wild-type. tPlus3a and tPlus3b positively or negatively regulated the expression of nearly 400 genes; tBRD-1 regulated 1,500 genes. Nearly 200 genes were regulated by both tPlus3a and tPlus3b and tBRD-1. tPlus3a and tPlus3b activated the Y-chromosomal genes kl-3 and kl-5, which indicates that tPlus3a and tPlus3b proteins are required for the function of distinct classes of genes. tPlus3a and tPlus3b and tBRD-1 repress genes relevant for seminal fluid and heat shock. We hypothesize that tPlus3a and tPlus3b proteins are required to specify the general transcriptional program in spermatocytes.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Fertility/genetics , Heat-Shock Proteins/metabolism , Y Chromosome/genetics , Animals , Dimerization , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Heat-Shock Proteins/chemistry , High-Throughput Nucleotide Sequencing , Male , RNA Interference , Sequence Analysis, RNA , Spermatocytes/metabolism , Transcription, Genetic
16.
Eur J Cell Biol ; 98(2-4): 103-115, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30679029

ABSTRACT

Spermiogenesis in Drosophila melanogaster is a highly conserved process and essential for male fertility. In this haploid phase of spermatogenesis, motile sperm are assembled from round cells, and flagella and needle-shaped nuclei with highly compacted genomes are formed. As transcription takes place mainly in spermatocytes and transcripts relevant for post-meiotic sperm development are translationally repressed for days, we comparatively analysed the proteome of larval testes (only germ cell stages before meiotic divisions), testes of 1-2-day-old pupae (germ cell stages before meiotic divisions, meiotic and early spermatid stages) and adult flies (germ cell stages before meiotic divisions, meiotic and early spermatid stages, late spermatids and sperm). We identified 6,171 proteins; 61 proteins were detected solely in one stage and are thus enriched, namely 34 in larval testes, 77 in pupal testes and 214 in adult testes. To substantiate our mass spectrometric data, we analysed the stage-specific synthesis and importance for male fertility of a number of uncharacterized proteins. For example, Mst84B (gene CG1988), a very basic cysteine- and lysine-rich nuclear protein and was present in the transition phase from a histone-based to a protamine-based chromatin structure. CG6332 encodes d-Theg, which is related to the mouse tHEG and human THEG proteins. Mutants of d-Theg were sterile due to the lack of sperm in the seminal vesicles. Our catalogue of proteins of the different stages of testis development in D. melanogaster will pave the road for future analyses of spermatogenesis.


Subject(s)
Drosophila Proteins/genetics , Infertility, Male/genetics , Proteome/genetics , Testis/metabolism , Animals , Drosophila Proteins/metabolism , Drosophila melanogaster , Male , Proteome/metabolism , Spermatogenesis/genetics , Testis/growth & development
17.
BMC Dev Biol ; 8: 68, 2008 Jun 27.
Article in English | MEDLINE | ID: mdl-18588663

ABSTRACT

BACKGROUND: Metal-responsive transcription factor 1 (MTF-1), which binds to metal response elements (MREs), plays a central role in transition metal detoxification and homeostasis. A Drosophila interactome analysis revealed two candidate dMTF-1 interactors, both of which are related to the small regulatory protein Dumpy-30 (Dpy-30) of the worm C. elegans. Dpy-30 is the founding member of a protein family involved in chromatin modifications, notably histone methylation. Mutants affect mating type in yeast and male mating in C. elegans. RESULTS: Constitutive expression of the stronger interactor, Dpy-30L1 (CG6444), in transgenic flies inhibits MTF-1 activity and results in elevated sensitivity to Cd(II) and Zn(II), an effect that could be rescued by co-overexpression of dMTF-1. Electrophoretic mobility shift assays (EMSA) suggest that Dpy-30L1 interferes with the binding of MTF-1 to its cognate MRE binding site. Dpy-30L1 is expressed in the larval brain, gonads, imaginal discs, salivary glands and in the brain, testes, ovaries and salivary glands of adult flies. Expression of the second interactor, Dpy-30L2 (CG11591), is restricted to larval male gonads, and to the testes of adult males. Consistent with these findings, dpy-30-like transcripts are also prominently expressed in mouse testes. Targeted gene disruption by homologous recombination revealed that dpy-30L1 knockout flies are viable and show no overt disruption of metal homeostasis. In contrast, the knockout of the male-specific dpy-30L2 gene results in male sterility, as does the double knockout of dpy-30L1 and dpy-30L2. A closer inspection showed that Dpy-30L2 is expressed in elongated spermatids but not in early or mature sperm. Mutant sperm had impaired motility and failed to accumulate in sperm storage organs of females. CONCLUSION: Our studies help to elucidate the physiological roles of the Dumpy-30 proteins, which are conserved from yeast to humans and typically act in concert with other nuclear proteins to modify chromatin structure and gene expression. The results from these studies reveal an inhibitory effect of Dpy-30L1 on MTF-1 and an essential role for Dpy-30L2 in male fertility.


Subject(s)
DNA-Binding Proteins/physiology , Drosophila Proteins/physiology , Transcription Factors/physiology , Animals , Drosophila , Female , Male , Mice , Protein Binding , Response Elements , Spermatozoa/cytology , Tissue Distribution , Transcription Factor MTF-1
18.
Mol Cell Biol ; 25(14): 6165-77, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15988027

ABSTRACT

Chromatin condensation is a typical feature of sperm cells. During mammalian spermiogenesis, histones are first replaced by transition proteins and then by protamines, while little is known for Drosophila melanogaster. Here we characterize three genes in the fly genome, Mst35Ba, Mst35Bb, and Mst77F. The results indicate that Mst35Ba and Mst35Bb encode dProtA and dProtB, respectively. These are considerably larger than mammalian protamines, but, as in mammals, both protamines contain typical cysteine/arginine clusters. Mst77F encodes a linker histone-like protein showing significant similarity to mammalian HILS1 protein. ProtamineA-enhanced green fluorescent protein (eGFP), ProtamineB-eGFP, and Mst77F-eGFP carrying Drosophila lines show that these proteins become the important chromosomal protein components of elongating spermatids, and His2AvDGFP vanishes. Mst77F mutants [ms(3)nc3] are characterized by small round nuclei and are sterile as males. These data suggest the major features of chromatin condensation in Drosophila spermatogenesis correspond to those in mammals. During early fertilization steps, the paternal pronucleus still contains protamines and Mst77F but regains a nucleosomal conformation before zygote formation. In eggs laid by sesame-deficient females, the paternal pronucleus remains in a protamine-based chromatin status but Mst77F-eGFP is removed, suggesting that the sesame gene product is essential for removal of protamines while Mst77F removal is independent of Sesame.


Subject(s)
Chromatin/metabolism , Drosophila Proteins/metabolism , Drosophila Proteins/physiology , Drosophila melanogaster/metabolism , Histones/metabolism , Protamines/metabolism , Spermatids/metabolism , Spermatogenesis/physiology , Amino Acid Sequence , Animals , Cell Cycle Proteins , Cell Nucleus/metabolism , Chromatin/genetics , Chromatin/physiology , Chromosomal Proteins, Non-Histone , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Female , Histone Chaperones , Histones/genetics , Infertility, Male/genetics , Male , Molecular Sequence Data , Mutation , Protamines/genetics , Spermatogenesis/genetics , Transcription Factors , Transcription, Genetic
19.
PLoS One ; 13(9): e0203622, 2018.
Article in English | MEDLINE | ID: mdl-30192860

ABSTRACT

Spermatogenesis in many species including Drosophila melanogaster is accompanied by major reorganisation of chromatin in post-meiotic stages, involving a nearly genome-wide displacement of histones by protamines, Mst77F and Protamine-like 99C. A proposed prerequisite for the histone-to-protamine transition is massive histone H4 hyper-acetylation prior to the switch. Here, we investigated the pattern of histone H3 lysine acetylation and general lysine crotonylation in D. melanogaster spermiogenesis to elucidate a possible role of these marks in chromatin reorganisation. Lysine crotonylation was strongest prior to remodelling and the deposition of this mark depended on the acetylation status of the spermatid chromatin. In contrast to H4 acetylation, individual H3 acetylation marks displayed surprisingly distinct patterns during the histone-to-protamine transition. We observed that Nejire, a histone acetyl transferase, is expressed during the time of histone-to-protamine transition. Nejire knock down led to strongly reduced fertility, which correlated with misshaped spermatid nuclei and a lack of mature sperm. protA and prtl99C transcript levels were reduced after knocking down Nejire. ProtB-eGFP, Mst77F-eGFP and Prtl99C-eGFP were synthesized at the late canoe stage, while histones were often not detectable. However, in some cysts histones persist in parallel to protamines. Therefore, we hypothesize that complete histone removal requires multiple histone modifications besides H3K18ac and H3K27ac. In summary, H3K18 and H3K27 acetylation during Drosophila spermatogenesis is dependent on Nejire or a yet uncharacterized acetyl transferase. We show that Nejire is required for male fertility since Nejire contributes to efficient transcription of protA and prtl99C, but not Mst77F, in spermatocytes, and to maturation of sperm.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , Histones/metabolism , p300-CBP Transcription Factors/metabolism , Acetylation , Animals , Drosophila Proteins/genetics , Fertility , Gene Expression Regulation , Gene Knockdown Techniques , Histones/genetics , Lysine/metabolism , Male , Protamines/genetics , Protamines/metabolism , Spermatogenesis , p300-CBP Transcription Factors/genetics
20.
Biol Open ; 6(12): 1876-1888, 2017 Dec 15.
Article in English | MEDLINE | ID: mdl-29122742

ABSTRACT

During Drosophila metamorphosis, nascent testis myotubes migrate from the prospective seminal vesicle of the genital disc onto pupal testes and then further to cover the testes with multinucleated smooth-like muscles. Here we show that DWnt2 is likely required for determination of testis-relevant myoblasts on the genital disc. Knock down of fibroblast growth factor receptor (FGFR) heartless by RNAi and a dominant-negative version revealed multiple functions of Heartless, namely regulation of the amount of myoblasts on the genital disc, connection of seminal vesicles and testes, and migration of muscles along the testes. Live imaging indicated that the downstream effector Stumps is required for migration of testis myotubes on the testis towards the apical tip. After myoblast fusion, myosin II is needed for migration of nascent testis myotubes, in which Thisbe-dependent fibroblast growth factor (FGF) signaling is activated. Cadherin-N is essential for connecting these single myofibers and for creating a firm testis muscle sheath that shapes and stabilizes the testis tubule. Based on these results, we propose a model for the migration of testis myotubes in which nascent testis myotubes migrate as a collective onto and along the testis, dependent on FGF-regulated expression of myosin II.

SELECTION OF CITATIONS
SEARCH DETAIL