Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Development ; 145(24)2018 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-30470702

RESUMO

The development of highly differentiated sperm cells that are specialized for navigating to and fusing with an oocyte is essential for sexual reproduction. As a major part of differentiation, sperm undergo extensive post-meiotic maturation en route to the oocyte. This is regulated largely by soma-derived cues. In Caenorhabditiselegans, this process is called sperm activation, and it transforms immotile spermatids into migratory fertilization-competent cells. Here, we show that the negative regulator of sperm activation, SWM-1, is produced in an unexpected cell type: body wall muscle. SWM-1 is secreted into the body cavity and enters the gonad; there, it is present with its likely target, TRY-5, a spermiogenesis activator. We show that, in addition to SWM-1, the somatic gonad and body fluid can exchange other factors, suggesting that soma-germ line transfer could affect other reproductive processes. In addition, we show that SWM-1 may have a separate role in the sperm migratory environment, to which it is contributed by both males and hermaphrodites. These findings reveal that late stages in gamete differentiation can be regulated at the whole-organism level by broadly secreted factors.This article has an associated 'The people behind the papers' interview.


Assuntos
Caenorhabditis elegans/fisiologia , Células Germinativas/fisiologia , Músculos/fisiologia , Motilidade dos Espermatozoides/fisiologia , Animais , Proteínas de Caenorhabditis elegans/metabolismo , Fertilidade , Genes Reporter , Organismos Hermafroditas/fisiologia , Masculino , Reprodução , Glândulas Seminais/metabolismo , Espermatozoides/fisiologia
2.
Dev Biol ; 436(2): 75-83, 2018 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-29477340

RESUMO

Successful fertilization requires that sperm are activated prior to contacting an oocyte. In C. elegans, this activation process, called spermiogenesis, transforms round immobile spermatids into motile, fertilization-competent spermatozoa. We describe the phenotypic and genetic characterization of spe-43, a new component of the spe-8 pathway, which is required for spermiogenesis in hermaphrodites; spe-43 hermaphrodites are self-sterile, while spe-43 males show wild-type fertility. When exposed to Pronase to activate sperm in vitro, spe-43 spermatids form long rigid spikes radiating outward from the cell periphery instead of forming a motile pseudopod, indicating that spermiogenesis initiates but is not completed. Using a combination of recombinant and deletion mapping and whole genome sequencing, we identified F09E8.1 as spe-43. SPE-43 is predicted to exist in two isoforms; one isoform appears to be a single-pass transmembrane protein while the other is predicted to be a secreted protein. SPE-43 can bind to other known sperm proteins, including SPE-4 and SPE-29, which are known to impact spermiogenesis. In summary, we have identified a membrane protein that is present in C. elegans sperm and is required for sperm activation via the hermaphrodite activation signal.


Assuntos
Proteínas de Caenorhabditis elegans/genética , Espermatogênese/genética , Espermatozoides/metabolismo , Animais , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Fertilidade/genética , Masculino , Mutação , Fenótipo , Polimorfismo de Nucleotídeo Único , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Espermatogênese/fisiologia , Espermatozoides/fisiologia , Sequenciamento Completo do Genoma
3.
Proc Natl Acad Sci U S A ; 114(33): 8806-8811, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28760991

RESUMO

Apoptotic cells undergo a series of morphological changes. These changes are dependent on caspase cleavage of downstream targets, but which targets are significant and how they facilitate the death process are not well understood. In Caenorhabditis elegans an increase in the refractility of the dying cell is a hallmark morphological change that is caspase dependent. We identify a presumptive transient receptor potential (TRP) cation channel, CED-11, that acts in the dying cell to promote the increase in apoptotic cell refractility. CED-11 is required for multiple other morphological changes during apoptosis, including an increase in electron density as visualized by electron microscopy and a decrease in cell volume. In ced-11 mutants, the degradation of apoptotic cells is delayed. Mutation of ced-11 does not cause an increase in cell survival but can enhance cell survival in other cell-death mutants, indicating that ced-11 facilitates the death process. In short, ced-11 acts downstream of caspase activation to promote the shrinkage, death, and degradation of apoptotic cells.


Assuntos
Apoptose/fisiologia , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Caspases/metabolismo , Tamanho Celular , Canais de Potencial de Receptor Transitório/metabolismo , Animais , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Caspases/genética , Canais de Potencial de Receptor Transitório/genética
4.
Worm ; 4(1): e1003002, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26430556

RESUMO

Sperm from the nematode C. elegans gain motility during a process termed activation, which they initiate in response to specific environmental signals. During this process, a number of subcellular rearrangements occur, culminating in an altered morphology that allows the cell to crawl toward and fertilize oocytes. Both hermaphrodites and males produce sperm, and redundant, sex-biased pathways regulate the sperm's activation. The male-derived signal for sperm activation involves TRY-5, a trypsin-like serine protease in seminal fluid, but until recently it was unknown what factors were active downstream of TRY-5. In our recent paper, we reported the discovery of SNF-10, a solute carrier 6 (SLC6) family protein that is expressed by sperm and connects the activation signal to changes in sperm morphology and, ultimately, the onset of motility. Here, we review our recent results, focusing on potential models for SNF-10's function in C. elegans, and additionally discuss the role SLC6 transporters may play in male reproductive biology from invertebrates to mammals.

5.
Elife ; 42015 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-25789512

RESUMO

Competition among sperm to fertilize oocytes is a ubiquitous feature of sexual reproduction as well as a profoundly important aspect of sexual selection. However, little is known about the cellular mechanisms sperm use to gain competitive advantage or how these mechanisms are regulated genetically. In this study, we utilize a forward genetic screen in Caenorhabditis elegans to identify a gene, comp-1, whose function is specifically required in competitive contexts. We show that comp-1 functions in sperm to modulate their migration through and localization within the reproductive tract, thereby promoting their access to oocytes. Contrary to previously described models, comp-1 mutant sperm show no defects in size or velocity, thereby defining a novel pathway for preferential usage. Our results indicate not only that sperm functional traits can influence the outcome of sperm competition, but also that these traits can be modulated in a context-dependent manner depending on the presence of competing sperm.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Espermatozoides/metabolismo , Quinases da Família src/metabolismo , Animais , Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/genética , Forma Celular , Tamanho Celular , Mapeamento Cromossômico , Cromossomos/genética , Feminino , Testes Genéticos , Organismos Hermafroditas/metabolismo , Masculino , Modelos Biológicos , Mutação/genética , Estrutura Terciária de Proteína , Pseudópodes/metabolismo , Motilidade dos Espermatozoides , Espermatozoides/citologia , Quinases da Família src/química , Quinases da Família src/genética
6.
Dev Biol ; 393(1): 171-82, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-24929237

RESUMO

Motility of sperm is crucial for their directed migration to the egg. The acquisition and modulation of motility are regulated to ensure that sperm move when and where needed, thereby promoting reproductive success. One specific example of this phenomenon occurs during differentiation of the ameboid sperm of Caenorhabditis elegans as they activate from a round spermatid to a mature, crawling spermatozoon. Sperm activation is regulated by redundant pathways to occur at a specific time and place for each sex. Here, we report the identification of the solute carrier 6 (SLC6) transporter protein SNF-10 as a key regulator of C. elegans sperm activation in response to male protease activation signals. We find that SNF-10 is present in sperm and is required for activation by the male but not by the hermaphrodite. Loss of both snf-10 and a hermaphrodite activation factor render sperm completely insensitive to activation. Using in vitro assays, we find that snf-10 mutant sperm show a specific deficit in response to protease treatment but not to other activators. Prior to activation, SNF-10 is present in the plasma membrane, where it represents a strong candidate to receive signals that lead to subcellular morphogenesis. After activation, it shows polarized localization to the cell body region that is dependent on membrane fusions mediated by the dysferlin FER-1. Our discovery of snf-10 offers insight into the mechanisms differentially employed by the two sexes to accomplish the common goal of producing functional sperm, as well as how the physiology of nematode sperm may be regulated to control motility as it is in mammals.


Assuntos
Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/fisiologia , Proteínas da Membrana Plasmática de Transporte de GABA/fisiologia , Motilidade dos Espermatozoides/fisiologia , Espermatozoides/citologia , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/biossíntese , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas da Membrana Plasmática de Transporte de GABA/biossíntese , Proteínas da Membrana Plasmática de Transporte de GABA/genética , Organismos Hermafroditas , Masculino , Proteínas de Membrana/metabolismo , Morfogênese , Mutação , Motilidade dos Espermatozoides/genética , Espermatogênese
7.
Semin Cell Dev Biol ; 29: 17-30, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24718317

RESUMO

In the nematode C. elegans, both males and self-fertile hermaphrodites produce sperm. As a result, researchers have been able to use a broad range of genetic and genomic techniques to dissect all aspects of sperm development and function. Their results show that the early stages of spermatogenesis are controlled by transcriptional and translational processes, but later stages are dominated by protein kinases and phosphatases. Once spermatids are produced, they participate in many interactions with other cells - signals from the somatic gonad determine when sperm activate and begin to crawl, signals from the female reproductive tissues guide the sperm, and signals from sperm stimulate oocytes to mature and be ovulated. The sperm also show strong competitive interactions with other sperm and oocytes. Some of the molecules that mediate these processes have conserved functions in animal sperm, others are conserved proteins that have been adapted for new roles in nematode sperm, and some are novel proteins that provide insights into evolutionary change. The advent of new techniques should keep this system on the cutting edge of research in cellular and reproductive biology.


Assuntos
Caenorhabditis elegans/fisiologia , Espermatogênese/fisiologia , Espermatozoides/citologia , Animais , Caenorhabditis elegans/embriologia , Caenorhabditis elegans/genética , Desenvolvimento Embrionário/fisiologia , Feminino , Proteínas de Helminto/metabolismo , Masculino , Oócitos/fisiologia , Transdução de Sinais , Espermatogênese/genética , Espermatozoides/metabolismo
8.
Worm ; 1(3): 151-4, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24058840

RESUMO

Seminal fluid factors have been shown to play a significant role in fertility in many animals. However, little is known about the contributions of seminal fluid to male fertility in C. elegans. In this commentary, we summarize our recent finding of a seminal fluid sperm activator, the serine protease TRY-5. TRY-5 is required for males to activate sperm, yet surprisingly it is not required for male fertility, likely due to redundancy with an activator present in hermaphrodites. TRY-5 is transferred to hermaphrodites during mating in a series of distinct release events just prior to transfer of sperm. Thus, we propose a model in which TRY-5 cleaves sperm cell surface proteins to trigger sperm maturation. We discuss other possible roles for seminal fluid factors in C. elegans and prospects for using TRY-5 as a marker for studies of male mating behavior and seminal fluid secretion.

9.
PLoS Genet ; 7(11): e1002375, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22125495

RESUMO

Seminal fluid proteins have been shown to play important roles in male reproductive success, but the mechanisms for this regulation remain largely unknown. In Caenorhabditis elegans, sperm differentiate from immature spermatids into mature, motile spermatozoa during a process termed sperm activation. For C. elegans males, sperm activation occurs during insemination of the hermaphrodite and is thought to be mediated by seminal fluid, but the molecular nature of this activity has not been previously identified. Here we show that TRY-5 is a seminal fluid protease that is required in C. elegans for male-mediated sperm activation. We observed that TRY-5::GFP is expressed in the male somatic gonad and is transferred along with sperm to hermaphrodites during mating. In the absence of TRY-5, male seminal fluid loses its potency to transactivate hermaphrodite sperm. However, TRY-5 is not required for either hermaphrodite or male fertility, suggesting that hermaphrodite sperm are normally activated by a distinct hermaphrodite-specific activator to which male sperm are also competent to respond. Within males, TRY-5::GFP localization within the seminal vesicle is antagonized by the protease inhibitor SWM-1. Together, these data suggest that TRY-5 functions as an extracellular activator of C. elegans sperm. The presence of TRY-5 within the seminal fluid couples the timing of sperm activation to that of transfer of sperm into the hermaphrodite uterus, where motility must be rapidly acquired. Our results provide insight into how C. elegans has adopted sex-specific regulation of sperm motility to accommodate its male-hermaphrodite mode of reproduction.


Assuntos
Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/enzimologia , Peptídeo Hidrolases/genética , Sêmen/enzimologia , Serina Proteases/genética , Espermatozoides/enzimologia , Animais , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Transtornos do Desenvolvimento Sexual/genética , Feminino , Fertilidade/genética , Regulação da Expressão Gênica , Gônadas/metabolismo , Masculino , Mutação , Peptídeo Hidrolases/metabolismo , Serina Proteases/metabolismo
10.
Curr Biol ; 16(3): 252-63, 2006 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-16461278

RESUMO

BACKGROUND: Sexual reproduction in animals requires the production of highly specialized motile sperm cells that can navigate to and fertilize ova. During sperm differentiation, nonmotile spermatids are remodeled into motile spermatozoa through a process known as spermiogenesis. In nematodes, spermiogenesis, or sperm activation, involves a rapid cellular morphogenesis that converts unpolarized round spermatids into polarized amoeboid spermatozoa capable of both motility and fertilization. RESULTS: Here we demonstrate, by genetic analysis and in vivo and in vitro cell-based assays, that the temporal and spatial localization of spermiogenesis are critical determinants of male fertility in C. elegans, a male/hermaphrodite species. We identify swm-1 as a factor important for male but not hermaphrodite fertility. We show that whereas in wild-type males, activation occurs after spermatids are transferred to the hermaphrodite, swm-1 mutants exhibit ectopic activation of sperm within the male reproductive tract. This ectopic activation leads to infertility by impeding sperm transfer. The SWM-1 protein is composed of a signal sequence and two trypsin inhibitor-like domains and likely functions as a secreted serine protease inhibitor that targets two distinct proteases. CONCLUSIONS: These findings support a model in which (1) proteolysis acts as an important in vivo trigger for sperm activation and (2) regulating the timing of proteolysis-triggered activation is crucial for male reproductive success. Furthermore, our data provide insight into how a common program of gamete differentiation can be modulated to allow males to participate in reproduction in the context of a male/hermaphrodite species where the capacity for hermaphrodite self-fertilization has rendered them nonessential for progeny production.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/fisiologia , Fertilidade/fisiologia , Inibidores de Serina Proteinase/metabolismo , Espermatogênese/fisiologia , Espermatozoides/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Movimento Celular/fisiologia , Transtornos do Desenvolvimento Sexual/metabolismo , Genitália Masculina/metabolismo , Masculino , Microscopia de Fluorescência , Modelos Biológicos , Dados de Sequência Molecular , Compostos Orgânicos , Análise de Sequência de DNA , Inibidores de Serina Proteinase/genética , Supressão Genética/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA