Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
PLoS Genet ; 17(9): e1009787, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34478447

RESUMEN

Comparative genomics has enabled the identification of genes that potentially evolved de novo from non-coding sequences. Many such genes are expressed in male reproductive tissues, but their functions remain poorly understood. To address this, we conducted a functional genetic screen of over 40 putative de novo genes with testis-enriched expression in Drosophila melanogaster and identified one gene, atlas, required for male fertility. Detailed genetic and cytological analyses showed that atlas is required for proper chromatin condensation during the final stages of spermatogenesis. Atlas protein is expressed in spermatid nuclei and facilitates the transition from histone- to protamine-based chromatin packaging. Complementary evolutionary analyses revealed the complex evolutionary history of atlas. The protein-coding portion of the gene likely arose at the base of the Drosophila genus on the X chromosome but was unlikely to be essential, as it was then lost in several independent lineages. Within the last ~15 million years, however, the gene moved to an autosome, where it fused with a conserved non-coding RNA and evolved a non-redundant role in male fertility. Altogether, this study provides insight into the integration of novel genes into biological processes, the links between genomic innovation and functional evolution, and the genetic control of a fundamental developmental process, gametogenesis.


Asunto(s)
Cromatina/metabolismo , Drosophila melanogaster/genética , Evolución Molecular , Espermátides/metabolismo , Animales , Núcleo Celular/metabolismo , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Fertilidad/genética , Masculino , Interferencia de ARN , Espermatogénesis/genética
2.
Mol Reprod Dev ; 87(1): 3-6, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31736234

RESUMEN

Molecular Reproduction and Development is delighted to announce that editorial board member Mariana F. Wolfner has been elected to the National Academy of Sciences. Here, Dr Wolfner is interviewed by two of her former postdocs. She discusses her path to studying reproduction and her career as a researcher and mentor.


Asunto(s)
Tutoría/métodos , Mentores/psicología , Investigadores/psicología , Animales , Investigación Biomédica/métodos , Drosophila/embriología , Drosophila/genética , Femenino , Humanos , National Academy of Sciences, U.S. , Reproducción/genética , Procesos de Determinación del Sexo/genética , Estados Unidos
3.
J Evol Biol ; 33(5): 629-641, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31991034

RESUMEN

Successful reproduction depends on interactions between numerous proteins beyond those involved directly in gamete fusion. Although such reproductive proteins evolve in response to sexual selection pressures, how networks of interacting proteins arise and evolve as reproductive phenotypes change remains an open question. Here, we investigated the molecular evolution of the 'sex peptide network' of Drosophila melanogaster, a functionally well-characterized reproductive protein network. In this species, the peptide hormone sex peptide (SP) and its interacting proteins cause major changes in female physiology and behaviour after mating. In contrast, females of more distantly related Drosophila species do not respond to SP. In spite of these phenotypic differences, we detected orthologs of all network proteins across 22 diverse Drosophila species and found evidence that most orthologs likely function in reproduction throughout the genus. Within SP-responsive species, we detected the recurrent, adaptive evolution of several network proteins, consistent with sexual selection acting to continually refine network function. We also found some evidence for adaptive evolution of several proteins along two specific phylogenetic lineages that correspond with increased expression of the SP receptor in female reproductive tracts or increased sperm length, respectively. Finally, we used gene expression profiling to examine the likely degree of functional conservation of the paralogs of an SP network protein that arose via gene duplication. Our results suggest a dynamic history for the SP network in which network members arose before the onset of robust SP-mediated responses and then were shaped by both purifying and positive selection.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Evolución Molecular , Péptidos y Proteínas de Señalización Intercelular/genética , Receptores de Péptidos/genética , Selección Sexual , Adaptación Biológica , Animales , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Femenino , Duplicación de Gen , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Masculino , Receptores de Péptidos/metabolismo , Reproducción/genética , Serina Proteasas/genética , Serina Proteasas/metabolismo
4.
Mol Biol Evol ; 34(5): 1066-1082, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28104747

RESUMEN

New genes arise through a variety of mechanisms, including the duplication of existing genes and the de novo birth of genes from noncoding DNA sequences. While there are numerous examples of duplicated genes with important functional roles, the functions of de novo genes remain largely unexplored. Many newly evolved genes are expressed in the male reproductive tract, suggesting that these evolutionary innovations may provide advantages to males experiencing sexual selection. Using testis-specific RNA interference, we screened 11 putative de novo genes in Drosophila melanogaster for effects on male fertility and identified two, goddard and saturn, that are essential for spermatogenesis and sperm function. Goddard knockdown (KD) males fail to produce mature sperm, while saturn KD males produce few sperm, and these function inefficiently once transferred to females. Consistent with a de novo origin, both genes are identifiable only in Drosophila and are predicted to encode proteins with no sequence similarity to any annotated protein. However, since high levels of divergence prevented the unambiguous identification of the noncoding sequences from which each gene arose, we consider goddard and saturn to be putative de novo genes. Within Drosophila, both genes have been lost in certain lineages, but show conserved, male-specific patterns of expression in the species in which they are found. Goddard is consistently found in single-copy and evolves under purifying selection. In contrast, saturn has diversified through gene duplication and positive selection. These data suggest that de novo genes can acquire essential roles in male reproduction.


Asunto(s)
Drosophila melanogaster/genética , Fertilidad/genética , Espermatogénesis/genética , Animales , Evolución Biológica , Drosophila/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Evolución Molecular , Duplicación de Gen/genética , Técnicas de Silenciamiento del Gen/métodos , Masculino , Espermatozoides/metabolismo , Testículo/metabolismo
5.
PLoS Genet ; 10(1): e1004108, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24453993

RESUMEN

Seminal fluid proteins transferred from males to females during copulation are required for full fertility and can exert dramatic effects on female physiology and behavior. In Drosophila melanogaster, the seminal protein sex peptide (SP) affects mated females by increasing egg production and decreasing receptivity to courtship. These behavioral changes persist for several days because SP binds to sperm that are stored in the female. SP is then gradually released, allowing it to interact with its female-expressed receptor. The binding of SP to sperm requires five additional seminal proteins, which act together in a network. Hundreds of uncharacterized male and female proteins have been identified in this species, but individually screening each protein for network function would present a logistical challenge. To prioritize the screening of these proteins for involvement in the SP network, we used a comparative genomic method to identify candidate proteins whose evolutionary rates across the Drosophila phylogeny co-vary with those of the SP network proteins. Subsequent functional testing of 18 co-varying candidates by RNA interference identified three male seminal proteins and three female reproductive tract proteins that are each required for the long-term persistence of SP responses in females. Molecular genetic analysis showed the three new male proteins are required for the transfer of other network proteins to females and for SP to become bound to sperm that are stored in mated females. The three female proteins, in contrast, act downstream of SP binding and sperm storage. These findings expand the number of seminal proteins required for SP's actions in the female and show that multiple female proteins are necessary for the SP response. Furthermore, our functional analyses demonstrate that evolutionary rate covariation is a valuable predictive tool for identifying candidate members of interacting protein networks.


Asunto(s)
Drosophila melanogaster/genética , Péptidos/genética , Reproducción/genética , Proteínas de Plasma Seminal/genética , Conducta Sexual Animal , Animales , Copulación , Drosophila melanogaster/fisiología , Femenino , Fertilidad/genética , Masculino , Oviposición/genética , Péptidos/metabolismo , Proteínas de Plasma Seminal/aislamiento & purificación , Proteínas de Plasma Seminal/metabolismo , Espermatozoides/metabolismo
6.
Mol Biol Evol ; 31(6): 1554-67, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24682282

RESUMEN

Gene duplication is an important mechanism for the evolution of new reproductive proteins. However, in most cases, each resulting paralog continues to function within the same sex. To investigate the possibility that seminal fluid proteins arise through duplicates of female reproductive genes that become "co-opted" by males, we screened female reproductive genes in Drosophila melanogaster for cases of duplication in which one of the resulting paralogs produces a protein in males that is transferred to females during mating. We identified a set of three tandemly duplicated genes that encode secreted serine-type endopeptidase homologs, two of which are expressed primarily in the female reproductive tract (RT), whereas the third is expressed specifically in the male RT and encodes a seminal fluid protein. Evolutionary and gene expression analyses across Drosophila species suggest that this family arose from a single-copy gene that was female-specific; after duplication, one paralog evolved male-specific expression. Functional tests of knockdowns of each gene in D. melanogaster show that one female-expressed gene is essential for full fecundity, and both female-expressed genes contribute singly or in combination to a female's propensity to remate. In contrast, knockdown of the male-expressed paralog had no significant effect on female fecundity or remating. These data are consistent with a model in which members of this gene family exert effects on females by acting on a common, female-expressed target. After duplication and male co-option of one paralog, the evolution of the interacting proteins could have resulted in differential strengths or effects of each paralog.


Asunto(s)
Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Duplicación de Gen , Genes de Insecto , Animales , Evolución Molecular , Femenino , Fertilidad/genética , Expresión Génica , Masculino , Modelos Genéticos , Familia de Multigenes , Filogenia , Reproducción/genética , Caracteres Sexuales
7.
bioRxiv ; 2024 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-39149251

RESUMEN

New genes arise through a variety of evolutionary processes and provide raw material for adaptation in the face of both natural and sexual selection. De novo evolved genes emerge from previously non-protein-coding DNA sequences, and many such genes are expressed in male reproductive structures. In Drosophila melanogaster, several putative de novo genes have evolved essential roles in spermatogenesis, but whether such genes can also impact sperm function beyond the male has not been investigated. We identified a putative de novo gene, katherine johnson (kj), that is required for high levels of male fertility. Males that do not express kj produce and transfer sperm that are stored normally in females, but sperm from these males enter eggs with severely reduced efficiency. Using a tagged transgenic rescue construct, we observed that KJ protein localizes to the nuclear periphery in various stages of spermatogenesis, but is not detectable in mature sperm. These data suggest that kj exerts an effect on sperm development, the loss of which results in reduced fertilization ability. While previous bioinformatic analyses suggested the kj gene was restricted to the melanogaster group of Drosophila, we identified putative orthologs with conserved synteny, male-biased expression, and predicted protein features across the genus, as well as instances of gene loss in some lineages. Thus, kj potentially arose in the Drosophila common ancestor and subsequently evolved an essential role in D. melanogaster. Our results demonstrate a new aspect of male reproduction that has been shaped by new gene evolution and provide a molecular foothold for further investigating the mechanism of sperm entry into eggs in Drosophila.

8.
J Proteome Res ; 12(7): 3246-54, 2013 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-23713831

RESUMEN

We describe the use of a targeted proteomics approach, selected reaction monitoring (SRM) mass spectrometry, to detect and assess RNAi-mediated depletion or "knockdown" of specific proteins from human cells and from Drosophila flies. This label-free approach does not require any specific reagents to confirm the depletion of RNAi target protein(s) in unfractionated cell or whole organism extracts. The protocol described here is general, can be developed rapidly, and can be multiplexed to detect and measure multiple proteins at once. Furthermore, the methodology can be extended to any tandem mass spectrometer, making it widely accessible. This methodology will be applicable to a wide range of basic science and clinical questions where RNAi-mediated protein depletion needs to be verified, or where differences in relative abundance of target proteins need to be rapidly assessed between samples.


Asunto(s)
Proteínas/aislamiento & purificación , Proteómica/métodos , Interferencia de ARN , Animales , Línea Celular , Drosophila , Técnicas de Silenciamiento del Gen , Humanos , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Proteínas/genética , Espectrometría de Masas en Tándem
9.
bioRxiv ; 2023 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-36798169

RESUMEN

In Drosophila melanogaster and other insects, the seminal fluid proteins (SFPs) and male sex pheromones that enter the female with sperm during mating are essential for fertility and induce profound post-mating effects on female physiology and behavior. The SFPs in D. melanogaster and other taxa include several members of the large gene family known as odorant binding proteins (Obps). Previous work in Drosophila has shown that some Obp genes are highly expressed in the antennae and can mediate behavioral responses to odorants, potentially by binding and carrying these molecules to odorant receptors. These observations have led to the hypothesis that the seminal Obps might act as molecular carriers for pheromones or other compounds important for male fertility in the ejaculate, though functional evidence in any species is lacking. Here, we used RNAi and CRISPR/Cas9 generated mutants to test the role of the seven seminal Obps in D. melanogaster fertility and the post-mating response (PMR). We found that Obp56g is required for male fertility and the induction of the PMR, whereas the other six genes had no effect on fertility when mutated individually. Obp56g is expressed in the male's ejaculatory bulb, an important tissue in the reproductive tract that synthesizes components of the mating plug. We found males lacking Obp56g fail to form a mating plug in the mated female's reproductive tract, leading to ejaculate loss and reduced sperm storage. We also examined the evolutionary history of these seminal Obp genes, as several studies have documented rapid evolution and turnover of SFP genes across taxa. We found extensive lability in gene copy number and evidence of positive selection acting on two genes, Obp22a and Obp51a. Comparative RNAseq data from the male reproductive tract of multiple Drosophila species revealed that Obp56g shows high male reproductive tract expression only in species of the melanogaster and obscura groups, though conserved head expression in all species tested. Together, these functional and expression data suggest that Obp56g may have been co-opted for a reproductive function over evolutionary time.

10.
Elife ; 122023 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-38126735

RESUMEN

In Drosophila melanogaster and other insects, the seminal fluid proteins (SFPs) and male sex pheromones that enter the female with sperm during mating are essential for fertility and induce profound post-mating effects on female physiology. The SFPs in D. melanogaster and other taxa include several members of the large gene family known as odorant binding proteins (Obps). Work in Drosophila has shown that some Obp genes are highly expressed in the antennae and can mediate behavioral responses to odorants, potentially by binding and carrying these molecules to odorant receptors. These observations have led to the hypothesis that the seminal Obps might act as molecular carriers for pheromones or other compounds important for male fertility, though functional evidence in any species is lacking. Here, we used functional genetics to test the role of the seven seminal Obps in D. melanogaster fertility and the post-mating response (PMR). We found that Obp56g is required for male fertility and the induction of the PMR, whereas the other six genes are dispensable. We found males lacking Obp56g fail to form a mating plug in the mated female's reproductive tract, leading to ejaculate loss and reduced sperm storage, likely due to its expression in the male ejaculatory bulb. We also examined the evolutionary history of these seminal Obp genes, as several studies have documented rapid evolution and turnover of SFP genes across taxa. We found extensive lability in gene copy number and evidence of positive selection acting on two genes, Obp22a and Obp51a. Comparative RNAseq data from the male reproductive tract of multiple Drosophila species revealed that Obp56g shows high male reproductive tract expression in a subset of taxa, though conserved head expression across the phylogeny. Together, these functional and expression data suggest that Obp56g may have been co-opted for a reproductive function over evolutionary time.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Masculino , Femenino , Animales , Drosophila melanogaster/fisiología , Odorantes , Proteínas de Drosophila/metabolismo , Semillas , Fertilidad/genética , Espermatozoides/fisiología , Conducta Sexual Animal
11.
Bioessays ; 32(1): 26-36, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20020477

RESUMEN

Reproductive proteins maintain species-specific barriers to fertilization, affect the outcome of sperm competition, mediate reproductive conflicts between the sexes, and potentially contribute to the formation of new species. However, the specific proteins and molecular mechanisms that underlie these processes are understood in only a handful of cases. Advances in genomic and proteomic technologies enable the identification of large suites of reproductive proteins, making it possible to dissect reproductive phenotypes at the molecular level. We first review these technological advances and describe how reproductive proteins are identified in diverse animal taxa. We then discuss the dynamic evolution of reproductive proteins and the potential selective forces that act on them. Finally, we describe molecular and genomic tools for functional analysis and detail how evolutionary data may be used to make predictions about interactions among reproductive proteins.


Asunto(s)
Evolución Molecular , Proteínas/genética , Proteínas/fisiología , Reproducción/genética , Reproducción/fisiología , Animales , Femenino , Duplicación de Gen , Variación Genética , Genitales Femeninos/fisiología , Genitales Masculinos/fisiología , Genómica , Humanos , Masculino , Espectrometría de Masas , Modelos Genéticos , Mapeo Peptídico , Fenotipo , Filogenia , Mapeo de Interacción de Proteínas , Proteómica , Selección Genética
12.
BMC Genomics ; 12: 306, 2011 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-21663664

RESUMEN

BACKGROUND: Seminal fluid plays an important role in successful fertilization, but knowledge of the full suite of proteins transferred from males to females during copulation is incomplete. The list of ejaculated proteins remains particularly scant in one of the best-studied mammalian systems, the house mouse (Mus domesticus), where artificial ejaculation techniques have proven inadequate. Here we investigate an alternative method for identifying ejaculated proteins, by isotopically labeling females with 15N and then mating them to unlabeled, vasectomized males. Proteins were then isolated from mated females and identified using mass spectrometry. In addition to gaining insights into possible functions and fates of ejaculated proteins, our study serves as proof of concept that isotopic labeling is a powerful means to study reproductive proteins. RESULTS: We identified 69 male-derived proteins from the female reproductive tract following copulation. More than a third of all spectra detected mapped to just seven genes known to be structurally important in the formation of the copulatory plug, a hard coagulum that forms shortly after mating. Seminal fluid is significantly enriched for proteins that function in protection from oxidative stress and endopeptidase inhibition. Females, on the other hand, produce endopeptidases in response to mating. The 69 ejaculated proteins evolve significantly more rapidly than other proteins that we previously identified directly from dissection of the male reproductive tract. CONCLUSION: Our study attempts to comprehensively identify the proteins transferred from males to females during mating, expanding the application of isotopic labeling to mammalian reproductive genomics. This technique opens the way to the targeted monitoring of the fate of ejaculated proteins as they incubate in the female reproductive tract.


Asunto(s)
Proteínas/análisis , Semen/metabolismo , Animales , Endopeptidasas/química , Endopeptidasas/metabolismo , Femenino , Marcaje Isotópico , Masculino , Ratones , Estrés Oxidativo , Proteínas/genética , Reproducción/genética , Semen/enzimología , Espectrometría de Masas en Tándem
13.
PLoS Biol ; 6(7): e178, 2008 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-18666829

RESUMEN

Across diverse taxa, seminal fluid proteins (Sfps) transferred at mating affect the reproductive success of both sexes. Such reproductive proteins often evolve under positive selection between species; because of this rapid divergence, Sfps are hypothesized to play a role in speciation by contributing to reproductive isolation between populations. In Drosophila, individual Sfps have been characterized and are known to alter male sperm competitive ability and female post-mating behavior, but a proteomic-scale view of the transferred Sfps has been missing. Here we describe a novel proteomic method that uses whole-organism isotopic labeling to detect transferred Sfps in mated female D. melanogaster. We identified 63 proteins, which were previously unknown to function in reproduction, and confirmed the transfer of dozens of predicted Sfps. Relative quantification of protein abundance revealed that several of these novel Sfps are abundant in seminal fluid. Positive selection and tandem gene duplication are the prevailing forces of Sfp evolution, and comparative proteomics with additional species revealed lineage-specific changes in seminal fluid content. We also report a proteomic-based gene discovery method that uncovered 19 previously unannotated genes in D. melanogaster. Our results demonstrate an experimental method to identify transferred proteins in any system that is amenable to isotopic labeling, and they underscore the power of combining proteomic and evolutionary analyses to shed light on the complex process of Drosophila reproduction.


Asunto(s)
Proteínas de Drosophila/metabolismo , Proteómica/métodos , Proteínas de Plasma Seminal/metabolismo , Conducta Sexual Animal/fisiología , Animales , ADN Complementario/química , ADN Complementario/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiología , Evolución Molecular , Femenino , Expresión Génica , Masculino , Espectrometría de Masas , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas de Plasma Seminal/genética , Proteínas de Plasma Seminal/fisiología , Análisis de Secuencia de ADN
14.
Nat Commun ; 12(1): 1667, 2021 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-33712569

RESUMEN

Comparative genomic studies have repeatedly shown that new protein-coding genes can emerge de novo from noncoding DNA. Still unknown is how and when the structures of encoded de novo proteins emerge and evolve. Combining biochemical, genetic and evolutionary analyses, we elucidate the function and structure of goddard, a gene which appears to have evolved de novo at least 50 million years ago within the Drosophila genus. Previous studies found that goddard is required for male fertility. Here, we show that Goddard protein localizes to elongating sperm axonemes and that in its absence, elongated spermatids fail to undergo individualization. Combining modelling, NMR and circular dichroism (CD) data, we show that Goddard protein contains a large central α-helix, but is otherwise partially disordered. We find similar results for Goddard's orthologs from divergent fly species and their reconstructed ancestral sequences. Accordingly, Goddard's structure appears to have been maintained with only minor changes over millions of years.


Asunto(s)
Drosophila/genética , Evolución Molecular , Animales , Fertilidad/genética , Regulación del Desarrollo de la Expresión Génica , Genómica , Masculino , Simulación de Dinámica Molecular , Proteínas/metabolismo , Espermátides , Espermatozoides , Transgenes
15.
Mol Biol Evol ; 26(8): 1733-43, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19420050

RESUMEN

Male reproductive fitness is strongly affected by seminal fluid. In addition to interacting with the female environment, seminal fluid mediates important physiological characteristics of sperm, including capacitation and motility. In mammals, the male reproductive tract shows a striking degree of compartmentalization, with at least six distinct tissue types contributing material that is combined with sperm in an ejaculate. Although studies of whole ejaculates have been undertaken in some species, we lack a comprehensive picture of the specific proteins produced by different accessory tissues. Here, we perform proteomic investigations of six regions of the male reproductive tract in mice -- seminal vesicles, anterior prostate, dorsolateral prostate, ventral prostate, bulbourethral gland, and bulbourethral diverticulum. We identify 766 proteins that could be mapped to 506 unique genes and compare them with a high-quality human seminal fluid data set. We find that Gene Ontology functions of seminal proteins are largely conserved between mice and humans. By placing these data in an evolutionary framework, we show that seminal vesicle proteins have experienced a significantly higher rate of nonsynonymous substitution compared with the genome, which could be the result of adaptive evolution. In contrast, proteins from the other five tissues showed significantly lower nonsynonymous substitution, revealing a previously unappreciated level of evolutionary constraint acting on the majority of male reproductive proteins.


Asunto(s)
Evolución Molecular , Genitales Masculinos/química , Ratones , Proteómica , Semen/química , Proteínas de Plasma Seminal/análisis , Animales , Humanos , Masculino , Próstata/química , Proteínas de Plasma Seminal/genética , Vesículas Seminales/química
16.
Insect Biochem Mol Biol ; 102: 43-51, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30217614

RESUMEN

Seminal fluid proteins elicit several post-mating physiological changes in mated Drosophila melanogaster females. Some of these changes persist for over a week after mating because the seminal protein that causes these changes, the Sex Peptide (SP), binds to sperm that are stored in the female reproductive tract. SP's sperm binding is mediated by a network of at least eight seminal proteins. We show here that some of these network proteins (CG1656, CG1652, CG9997 and Antares) bind to sperm within 2 h of mating, like SP. However, while SP remains bound to sperm at 4 days post-mating, none of the other network proteins are detectable at this time. We also observed that the same network proteins are detectable at 2 h post-mating in seminal receptacle tissue from which sperm have been removed, but are no longer detectable there by 4 days post-mating, suggesting short-term retention of these proteins in this female sperm storage organ. Our results suggest that these network proteins act transiently to facilitate the conditions for SP's binding to sperm, perhaps by modifying SP or the sperm surface, but are not part of a long-acting complex that stably attaches SP to sperm.


Asunto(s)
Proteínas de Drosophila/metabolismo , Genitales Femeninos/fisiología , Proteínas de Plasma Seminal/metabolismo , Espermatozoides/metabolismo , Animales , Drosophila melanogaster , Femenino , Masculino , Factores de Tiempo
18.
Genome Res ; 19(5): 886-96, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19411605

RESUMEN

As genomic sequences become easier to acquire, shotgun proteomics will play an increasingly important role in genome annotation. With proteomics, researchers can confirm and revise existing genome annotations and discover completely new genes. Proteomic-based de novo gene discovery should be especially useful for sets of genes with characteristics that make them difficult to predict with gene-finding algorithms. Here, we report the proteomic discovery of 19 previously unannotated genes encoding seminal fluid proteins (Sfps) that are transferred from males to females during mating in Drosophila. Using bioinformatics, we detected putative orthologs of these genes, as well as 19 others detected by the same method in a previous study, across several related species. Gene expression analysis revealed that nearly all predicted orthologs are transcribed and that most are expressed in a male-specific or male-biased manner. We suggest several reasons why these genes escaped computational prediction. Like annotated Sfps, many of these new proteins show a pattern of adaptive evolution, consistent with their potential role in influencing male sperm competitive ability. However, in contrast to annotated Sfps, these new genes are shorter, have a higher rate of nonsynonymous substitution, and have a markedly lower GC content in coding regions. Our data demonstrate the utility of applying proteomic gene discovery methods to a specific biological process and provide a more complete picture of the molecules that are critical to reproductive success in Drosophila.


Asunto(s)
Biología Computacional/métodos , Drosophila/genética , Evolución Molecular , Genes de Insecto , Proteómica/métodos , Proteínas de Plasma Seminal/genética , Animales , Femenino , Duplicación de Gen , Masculino , Datos de Secuencia Molecular
19.
Mol Biol Evol ; 24(9): 2081-90, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17630281

RESUMEN

While gene duplication is a major source of evolutionary novelty, the importance of this process in reproductive protein evolution has not been widely investigated. Here, we report the first known case of gene duplication of abalone sperm lysin in an allopatric subspecies found in the Eastern Atlantic, Haliotis tuberculata coccinea. Mass spectrometry identified both copies of the lysin protein in testis tissue, and 3-dimensional structural modeling suggests that both proteins remain functional. We also detected positive selection acting on both paralogs after duplication and found evidence of a recent selective sweep. Because H. t. coccinea occurs in geographic isolation from other abalone species, these findings suggest that the evolution of lysin is not driven to create reproductive barriers to unfit hybrid formation with an overlapping species. Instead, sexual selection or sexual conflict acting during abalone fertilization could be responsible for the recent positive selection on this protein. The presence of multiple, rapidly evolving lysin genes in H. tuberculata presents an opportunity to study the early stages of diversification of a protein whose function is well understood.


Asunto(s)
Gastrópodos/genética , Duplicación de Gen , Mucoproteínas/genética , Selección Genética , Espermatozoides/metabolismo , Secuencia de Aminoácidos , Animales , Cromatografía Liquida , Electroforesis en Gel de Poliacrilamida , Gastrópodos/clasificación , Masculino , Modelos Moleculares , Datos de Secuencia Molecular , Mucoproteínas/clasificación , Mucoproteínas/metabolismo , Filogenia , Polimorfismo Genético , Conformación Proteica , Homología de Secuencia de Aminoácido , Espectrometría de Masas en Tándem
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA