Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 124
Filtrar
1.
Mol Cell ; 74(5): 1069-1085.e11, 2019 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-31000436

RESUMEN

Orderly segregation of chromosomes during meiosis requires that crossovers form between homologous chromosomes by recombination. Programmed DNA double-strand breaks (DSBs) initiate meiotic recombination. We identify ANKRD31 as a key component of complexes of DSB-promoting proteins that assemble on meiotic chromosome axes. Genome-wide, ANKRD31 deficiency causes delayed recombination initiation. In addition, loss of ANKRD31 alters DSB distribution because of reduced selectivity for sites that normally attract DSBs. Strikingly, ANKRD31 deficiency also abolishes uniquely high rates of recombination that normally characterize pseudoautosomal regions (PARs) of X and Y chromosomes. Consequently, sex chromosomes do not form crossovers, leading to chromosome segregation failure in ANKRD31-deficient spermatocytes. These defects co-occur with a genome-wide delay in assembling DSB-promoting proteins on autosome axes and loss of a specialized PAR-axis domain that is highly enriched for DSB-promoting proteins in wild type. Thus, we propose a model for spatiotemporal patterning of recombination by ANKRD31-dependent control of axis-associated DSB-promoting proteins.


Asunto(s)
Proteínas Portadoras/genética , Roturas del ADN de Doble Cadena , Recombinación Homóloga/genética , Meiosis/genética , Animales , Proteínas Portadoras/química , Segregación Cromosómica/genética , Masculino , Ratones , Regiones Pseudoautosómicas/genética , Espermatocitos/crecimiento & desarrollo , Espermatocitos/metabolismo , Cromosoma X/genética , Cromosoma Y/genética
2.
Development ; 148(10)2021 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-33998651

RESUMEN

Heterochromatin-related epigenetic mechanisms, such as DNA methylation, facilitate pairing of homologous chromosomes during the meiotic prophase of mammalian spermatogenesis. In pro-spermatogonia, de novo DNA methylation plays a key role in completing meiotic prophase and initiating meiotic division. However, the role of maintenance DNA methylation in the regulation of meiosis, especially in the adult, is not well understood. Here, we reveal that NP95 (also known as UHRF1) and DNMT1 - two essential proteins for maintenance DNA methylation - are co-expressed in spermatogonia and are necessary for meiosis in male germ cells. We find that Np95- or Dnmt1-deficient spermatocytes exhibit spermatogenic defects characterized by synaptic failure during meiotic prophase. In addition, assembly of pericentric heterochromatin clusters in early meiotic prophase, a phenomenon that is required for subsequent pairing of homologous chromosomes, is disrupted in both mutants. Based on these observations, we propose that DNA methylation, established in pre-meiotic spermatogonia, regulates synapsis of homologous chromosomes and, in turn, quality control of male germ cells. Maintenance DNA methylation, therefore, plays a role in ensuring faithful transmission of both genetic and epigenetic information to offspring.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/genética , Emparejamiento Cromosómico/genética , ADN (Citosina-5-)-Metiltransferasa 1/genética , Metilación de ADN/genética , Espermatocitos/crecimiento & desarrollo , Espermatogénesis/genética , Ubiquitina-Proteína Ligasas/genética , Células Madre Germinales Adultas/citología , Animales , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Epigénesis Genética/genética , Heterocromatina/metabolismo , Masculino , Ratones , Ratones Noqueados , Espermatocitos/fisiología , Espermatogénesis/fisiología , Ubiquitina-Proteína Ligasas/metabolismo
3.
Development ; 148(5)2021 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-33558389

RESUMEN

Many specialized cells use unconventional strategies of cytoskeletal control. Nematode spermatocytes discard their actin and tubulin following meiosis, and instead employ the regulated assembly/disassembly of the Major Sperm Protein (MSP) to drive sperm motility. However, prior to the meiotic divisions, MSP is sequestered through its assembly into paracrystalline structures called fibrous bodies (FBs). The accessory proteins that direct this sequestration process have remained mysterious. This study reveals SPE-18 as an intrinsically disordered protein that is essential for MSP assembly within FBs. In spe-18 mutant spermatocytes, MSP forms disorganized cortical fibers, and the cells arrest in meiosis without forming haploid sperm. In wild-type spermatocytes, SPE-18 localizes to pre-FB complexes and functions with the kinase SPE-6 to localize MSP assembly. Changing patterns of SPE-18 localization uncover previously unappreciated complexities in FB maturation. Later, within newly individualized spermatids, SPE-18 is rapidly lost, yet SPE-18 loss alone is insufficient for MSP disassembly. Our findings reveal an alternative strategy for sequestering cytoskeletal elements, not as monomers but in localized, bundled polymers. Additionally, these studies provide an important example of disordered proteins promoting ordered cellular structures.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Proteínas Intrínsecamente Desordenadas/metabolismo , Espermatocitos/metabolismo , Secuencia de Aminoácidos , Animales , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/genética , Puntos de Control del Ciclo Celular , Citoesqueleto/metabolismo , Proteínas Intrínsecamente Desordenadas/química , Proteínas Intrínsecamente Desordenadas/genética , Masculino , Meiosis , Mutagénesis , Alineación de Secuencia , Espermátides/metabolismo , Espermatocitos/citología , Espermatocitos/crecimiento & desarrollo , Espermatogénesis
4.
PLoS Genet ; 17(6): e1009655, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34181646

RESUMEN

During spermatogenesis, the process in which sperm for fertilization are produced from germline cells, gene expression is spatiotemporally highly regulated. In Drosophila, successful expression of extremely large male fertility factor genes on Y-chromosome spanning some megabases due to their gigantic intron sizes is crucial for spermatogenesis. Expression of such extremely large genes must be challenging, but the molecular mechanism that allows it remains unknown. Here we report that a novel RNA-binding protein Maca, which contains two RNA-recognition motifs, is crucial for this process. maca null mutant male flies exhibited a failure in the spermatid individualization process during spermatogenesis, lacked mature sperm, and were completely sterile, while maca mutant female flies were fully fertile. Proteomics and transcriptome analyses revealed that both protein and mRNA abundance of the gigantic male fertility factor genes kl-2, kl-3, and kl-5 (kl genes) are significantly decreased, where the decreases of kl-2 are particularly dramatic, in maca mutant testes. Splicing of the kl-3 transcripts was also dysregulated in maca mutant testes. All these physiological and molecular phenotypes were rescued by a maca transgene in the maca mutant background. Furthermore, we found that in the control genetic background, Maca is exclusively expressed in spermatocytes in testes and enriched at Y-loop A/C in the nucleus, where the kl-5 primary transcripts are localized. Our data suggest that Maca increases transcription processivity, promotes successful splicing of gigantic introns, and/or protects transcripts from premature degradation, of the kl genes. Our study identified a novel RNA-binding protein Maca that is crucial for successful expression of the gigantic male fertility factor genes, spermatogenesis, and male fertility.


Asunto(s)
Drosophila melanogaster/genética , Proteínas de Unión al ARN/genética , Espermátides/metabolismo , Espermatocitos/metabolismo , Espermatogénesis/genética , Transcriptoma , Animales , Drosophila melanogaster/citología , Drosophila melanogaster/metabolismo , Femenino , Fertilidad/genética , Regulación de la Expresión Génica , Ontología de Genes , Genes Reporteros , Prueba de Complementación Genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Masculino , Anotación de Secuencia Molecular , Mutación , Proteínas de Unión al ARN/metabolismo , Espermátides/citología , Espermátides/crecimiento & desarrollo , Espermatocitos/citología , Espermatocitos/crecimiento & desarrollo , Testículo/citología , Testículo/metabolismo , Cromosoma Y/química
5.
J Med Genet ; 58(4): 254-263, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32527956

RESUMEN

BACKGROUND: Mutation in S-phase cyclin A-associated protein rin the endoplasmic reticulum (SCAPER) have been found across ethnicities and have been shown to cause variable penetrance of an array of pathological traits, including intellectual disability, retinitis pigmentosa and ciliopathies. METHODS: Human clinical phenotyping, surgical testicular sperm extraction and testicular tissue staining. Generation and analysis of short spindle 3 (ssp3) (SCAPER orthologue) Drosophila CAS9-knockout lines. In vitro microtubule (MT) binding assayed by total internal reflection fluorescence microscopy. RESULTS: We show that patients homozygous for a SCAPER mutation lack SCAPER expression in spermatogonia (SPG) and are azoospermic due to early defects in spermatogenesis, leading to the complete absence of meiotic cells. Interestingly, Drosophila null mutants for the ubiquitously expressed ssp3 gene are viable and female fertile but male sterile. We further show that male sterility in ssp3 null mutants is due to failure in both chromosome segregation and cytokinesis. In cells undergoing male meiosis, the MTs emanating from the centrosomes do not appear to interact properly with the chromosomes, which remain dispersed within dividing spermatocytes (SPCs). In addition, mutant SPCs are unable to assemble a normal central spindle and undergo cytokinesis. Consistent with these results, an in vitro assay demonstrated that both SCAPER and Ssp3 directly bind MTs. CONCLUSIONS: Our results show that SCAPER null mutations block the entry into meiosis of SPG, causing azoospermia. Null mutations in ssp3 specifically disrupt MT dynamics during male meiosis, leading to sterility. Moreover, both SCAPER and Ssp3 bind MTs in vitro. These results raise the intriguing possibility of a common feature between human and Drosophila meiosis.


Asunto(s)
Proteínas Portadoras/genética , Infertilidad Masculina/genética , Microtúbulos/genética , Serina Endopeptidasas/genética , Animales , Segregación Cromosómica/genética , Modelos Animales de Enfermedad , Drosophila melanogaster/genética , Predisposición Genética a la Enfermedad , Humanos , Infertilidad Masculina/patología , Masculino , Meiosis/genética , Mutación/genética , Espermatocitos/crecimiento & desarrollo , Espermatocitos/patología , Huso Acromático/genética , Huso Acromático/patología , Testículo/crecimiento & desarrollo , Testículo/patología
6.
PLoS Genet ; 15(1): e1007730, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30653507

RESUMEN

Meiosis is a cellular program that generates haploid gametes for sexual reproduction. While chromosome events that contribute to reducing ploidy (homologous chromosome pairing, synapsis, and recombination) are well conserved, their execution varies across species and even between sexes of the same species. The telomere bouquet is a conserved feature of meiosis that was first described nearly a century ago, yet its role is still debated. Here we took advantage of the prominent telomere bouquet in zebrafish, Danio rerio, and super-resolution microscopy to show that axis morphogenesis, synapsis, and the formation of double-strand breaks (DSBs) all take place within the immediate vicinity of telomeres. We established a coherent timeline of events and tested the dependence of each event on the formation of Spo11-induced DSBs. First, we found that the axis protein Sycp3 loads adjacent to telomeres and extends inward, suggesting a specific feature common to all telomeres seeds the development of the axis. Second, we found that newly formed axes near telomeres engage in presynaptic co-alignment by a mechanism that depends on DSBs, even when stable juxtaposition of homologous chromosomes at interstitial regions is not yet evident. Third, we were surprised to discover that ~30% of telomeres in early prophase I engage in associations between two or more chromosome ends and these interactions decrease in later stages. Finally, while pairing and synapsis were disrupted in both spo11 males and females, their reproductive phenotypes were starkly different; spo11 mutant males failed to produce sperm while females produced offspring with severe developmental defects. Our results support zebrafish as an important vertebrate model for meiosis with implications for differences in fertility and genetically derived birth defects in males and females.


Asunto(s)
Cromosomas/genética , Endodesoxirribonucleasas/genética , Meiosis/genética , Telómero/genética , Animales , Emparejamiento Cromosómico/genética , Roturas del ADN de Doble Cadena , Desarrollo Embrionario/genética , Femenino , Hibridación Fluorescente in Situ , Masculino , Profase/genética , Espermatocitos/crecimiento & desarrollo , Espermatocitos/metabolismo , Testículo/crecimiento & desarrollo , Testículo/patología , Pez Cebra/genética
7.
PLoS Genet ; 15(6): e1008177, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31170160

RESUMEN

During meiotic prophase I, double-strand breaks (DSBs) initiate homologous recombination leading to non-crossovers (NCOs) and crossovers (COs). In mouse, 10% of DSBs are designated to become COs, primarily through a pathway dependent on the MLH1-MLH3 heterodimer (MutLγ). Mlh3 contains an endonuclease domain that is critical for resolving COs in yeast. We generated a mouse (Mlh3DN/DN) harboring a mutation within this conserved domain that is predicted to generate a protein that is catalytically inert. Mlh3DN/DN males, like fully null Mlh3-/- males, have no spermatozoa and are infertile, yet spermatocytes have grossly normal DSBs and synapsis events in early prophase I. Unlike Mlh3-/- males, mutation of the endonuclease domain within MLH3 permits normal loading and frequency of MutLγ in pachynema. However, key DSB repair factors (RAD51) and mediators of CO pathway choice (BLM helicase) persist into pachynema in Mlh3DN/DN males, indicating a temporal delay in repair events and revealing a mechanism by which alternative DSB repair pathways may be selected. While Mlh3DN/DN spermatocytes retain only 22% of wildtype chiasmata counts, this frequency is greater than observed in Mlh3-/- males (10%), suggesting that the allele may permit partial endonuclease activity, or that other pathways can generate COs from these MutLγ-defined repair intermediates in Mlh3DN/DN males. Double mutant mice homozygous for the Mlh3DN/DN and Mus81-/- mutations show losses in chiasmata close to those observed in Mlh3-/- males, indicating that the MUS81-EME1-regulated crossover pathway can only partially account for the increased residual chiasmata in Mlh3DN/DN spermatocytes. Our data demonstrate that mouse spermatocytes bearing the MLH1-MLH3DN/DN complex display the proper loading of factors essential for CO resolution (MutSγ, CDK2, HEI10, MutLγ). Despite these functions, mice bearing the Mlh3DN/DN allele show defects in the repair of meiotic recombination intermediates and a loss of most chiasmata.


Asunto(s)
Proteínas de Unión al ADN/genética , Endonucleasas/genética , Profase Meiótica I/genética , Proteínas MutL/genética , Animales , Emparejamiento Cromosómico/genética , Intercambio Genético , Roturas del ADN de Doble Cadena , Reparación del ADN/genética , Recombinación Homóloga/genética , Masculino , Meiosis/genética , Ratones , Homólogo 1 de la Proteína MutL/genética , Proteínas MutS/genética , Recombinasa Rad51/genética , Espermatocitos/crecimiento & desarrollo , Espermatocitos/metabolismo
8.
PLoS Genet ; 15(5): e1008028, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31071079

RESUMEN

Intron gigantism, where genes contain megabase-sized introns, is observed across species, yet little is known about its purpose or regulation. Here we identify a unique gene expression program utilized for the proper expression of genes with intron gigantism. We find that two Drosophila genes with intron gigantism, kl-3 and kl-5, are transcribed in a spatiotemporal manner over the course of spermatocyte differentiation, which spans ~90 hours. The introns of these genes contain megabases of simple satellite DNA repeats that comprise over 99% of the gene loci, and these satellite-DNA containing introns are transcribed. We identify two RNA-binding proteins that specifically localize to kl-3 and kl-5 transcripts and are needed for the successful transcription or processing of these genes. We propose that genes with intron gigantism require a unique gene expression program, which may serve as a platform to regulate gene expression during cellular differentiation.


Asunto(s)
ADN Satélite/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Intrones , Espermatocitos/metabolismo , Espermatogénesis/genética , Animales , ADN Satélite/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/metabolismo , Exones , Regulación del Desarrollo de la Expresión Génica , Masculino , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Transducción de Señal , Espermatocitos/citología , Espermatocitos/crecimiento & desarrollo , Transcripción Genética , Cromosoma Y/química
9.
Differentiation ; 114: 49-57, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32585553

RESUMEN

Long noncoding RNAs (lncRNAs) have been shown to execute key roles in spermatogenesis. However, little is known about how lncRNAs gene expression is itself regulated in the germ cells of testis. We previously demonstrated that high expression of lncRNA-Gm2044 exists in spermatocytes and can regulate male germ cell proliferation. Here, the transcriptional regulation of lnRNA-Gm2044 expression in spermatocytes and the downstream signaling were further explored. A bioinformatics assessment predicted two potential binding-sites for the spermatocyte-specific transcription factor A-MYB in the promoter region of lncRNA-Gm2044. Our results proved that the transcription factor A-MYB promotes the expression of lncRNA-Gm2044 in mouse spermatocyte-derived GC-2spd(ts) cells. ChIP and luciferase assays verified that A-MYB mainly binds to the distal promoter region (-819 bp relative to the transcription start site) of lncRNA-Gm2044 and regulates lncRNA-Gm2044 expression through the -819 bp binding-site. In addition, we confirmed that lncRNA-Gm2044 functions as a miR-335-3p sponge to enhance the levels of miR-335-3p's direct target protein, Sycp1. Furthermore, A-MYB can up-regulate Sycp1 expression and down-regulate GC-2spd(ts) cell proliferation by activating its target, lncRNA-Gm2044. Overexpression of lncRNA-Gm2044 or knockdown of miR-335-3p can, at least partially, rescue the effects of A-MYB on Sycp1 expression and GC-2spd(ts) cell proliferation.Taken together, our results provide new information on the mechanistic roles of lncRNA-miRNA in transcription factor A-MYB regulation of spermatocyte function.


Asunto(s)
Proteínas de Unión al ADN/genética , MicroARNs/genética , Proteínas Proto-Oncogénicas c-myb/genética , ARN Largo no Codificante/genética , Espermatocitos/citología , Transactivadores/genética , Animales , Diferenciación Celular/genética , Regulación del Desarrollo de la Expresión Génica/genética , Masculino , Ratones , Espermatocitos/crecimiento & desarrollo , Transcripción Genética/genética , Activación Transcripcional/genética
10.
PLoS Genet ; 13(7): e1006926, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28749988

RESUMEN

Mammalian genomes harbor millions of retrotransposon copies, some of which are transpositionally active. In mouse prospermatogonia, PIWI-interacting small RNAs (piRNAs) combat retrotransposon activity to maintain the genomic integrity. The piRNA system destroys retrotransposon-derived RNAs and guides de novo DNA methylation at some retrotransposon promoters. However, it remains unclear whether DNA methylation contributes to retrotransposon silencing in prospermatogonia. We have performed comprehensive studies of DNA methylation and polyA(+) RNAs (transcriptome) in developing male germ cells from Pld6/Mitopld and Dnmt3l knockout mice, which are defective in piRNA biogenesis and de novo DNA methylation, respectively. The Dnmt3l mutation greatly reduced DNA methylation levels at most retrotransposons, but its impact on their RNA abundance was limited in prospermatogonia. In Pld6 mutant germ cells, although only a few retrotransposons exhibited reduced DNA methylation, many showed increased expression at the RNA level. More detailed analysis of RNA sequencing, nascent RNA quantification, profiling of cleaved RNA ends, and the results obtained from double knockout mice suggest that PLD6 works mainly at the posttranscriptional level. The increase in retrotransposon expression was larger in Pld6 mutants than it was in Dnmt3l mutants, suggesting that RNA degradation by the piRNA system plays a more important role than does DNA methylation in prospermatogonia. However, DNA methylation had a long-term effect: hypomethylation caused by the Pld6 or Dnmt3l mutation resulted in increased retrotransposon expression in meiotic spermatocytes. Thus, posttranscriptional silencing plays an important role in the early stage of germ cell development, then transcriptional silencing becomes important in later stages. In addition, intergenic and intronic retrotransposon sequences, in particular those containing the antisense L1 promoters, drove ectopic expression of nearby genes in both mutant spermatocytes, suggesting that retrotransposon silencing is important for the maintenance of not only genomic integrity but also transcriptomic integrity.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN/genética , Células Germinativas/crecimiento & desarrollo , Proteínas Mitocondriales/genética , Fosfolipasa D/genética , Transcripción Genética , Animales , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Ratones Noqueados , Interferencia de ARN , Estabilidad del ARN/genética , ARN Interferente Pequeño/genética , Retroelementos/genética , Espermatocitos/crecimiento & desarrollo , Testículo/crecimiento & desarrollo , Testículo/metabolismo , Transcriptoma/genética
11.
PLoS Genet ; 13(6): e1006845, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28617799

RESUMEN

To protect germ cells from genomic instability, surveillance mechanisms ensure meiosis occurs properly. In mammals, spermatocytes that display recombination defects experience a so-called recombination-dependent arrest at the pachytene stage, which relies on the MRE11 complex-ATM-CHK2 pathway responding to unrepaired DNA double-strand breaks (DSBs). Here, we asked if p53 family members-targets of ATM and CHK2-participate in this arrest. We bred double-mutant mice combining a mutation of a member of the p53 family (p53, TAp63, or p73) with a Trip13 mutation. Trip13 deficiency triggers a recombination-dependent response that arrests spermatocytes in pachynema before they have incorporated the testis-specific histone variant H1t into their chromatin. We find that deficiency for either p53 or TAp63, but not p73, allowed spermatocytes to progress further into meiotic prophase despite the presence of numerous unrepaired DSBs. Even so, the double mutant spermatocytes apoptosed at late pachynema because of sex body deficiency; thus p53 and TAp63 are dispensable for arrest caused by sex body defects. These data affirm that recombination-dependent and sex body-deficient arrests occur via genetically separable mechanisms.


Asunto(s)
Meiosis/genética , Fosfoproteínas/genética , Recombinación Genética , Transactivadores/genética , Proteína p53 Supresora de Tumor/genética , Animales , Apoptosis/genética , Puntos de Control del Ciclo Celular , Cromatina/genética , Roturas del ADN de Doble Cadena , Reparación del ADN/genética , Histonas/genética , Masculino , Ratones , Fase Paquiteno/genética , Espermatocitos/crecimiento & desarrollo , Espermatocitos/metabolismo , Testículo/crecimiento & desarrollo , Testículo/metabolismo
12.
Proc Natl Acad Sci U S A ; 114(47): E10132-E10141, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29109271

RESUMEN

Mammalian spermatogenesis is an elaborately organized differentiation process, starting with diploid spermatogonia, which include germ-line stem cells, and ending with haploid spermatozoa. The process involves four pivotal transitions occurring in physical proximity: spermatogonial differentiation, meiotic initiation, initiation of spermatid elongation, and release of spermatozoa. We report how the four transitions are coordinated in mice. Two premeiotic transitions, spermatogonial differentiation and meiotic initiation, were known to be coregulated by an extrinsic signal, retinoic acid (RA). Our chemical manipulations of RA levels in mouse testes now reveal that RA also regulates the two postmeiotic transitions: initiation of spermatid elongation and spermatozoa release. We measured RA concentrations and found that they changed periodically, as also reflected in the expression patterns of an RA-responsive gene, STRA8; RA levels were low before the four transitions, increased when the transitions occurred, and remained elevated thereafter. We found that pachytene spermatocytes, which express an RA-synthesizing enzyme, Aldh1a2, contribute directly and significantly to RA production in testes. Indeed, chemical and genetic depletion of pachytene spermatocytes revealed that RA from pachytene spermatocytes was required for the two postmeiotic transitions, but not for the two premeiotic transitions. We conclude that the premeiotic transitions are coordinated by RA from Sertoli (somatic) cells. Once germ cells enter meiosis, pachytene spermatocytes produce RA to coordinate the two postmeiotic transitions. In combination, these elements underpin the spatiotemporal coordination of spermatogenesis and ensure its prodigious output in adult males.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Aldehído Deshidrogenasa/genética , Regulación del Desarrollo de la Expresión Génica , Espermatogénesis/genética , Espermatozoides/metabolismo , Tretinoina/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Aldehído Deshidrogenasa/metabolismo , Familia de Aldehído Deshidrogenasa 1 , Animales , Diferenciación Celular , Masculino , Meiosis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fase Paquiteno , Retinal-Deshidrogenasa , Transducción de Señal , Espermátides/citología , Espermátides/crecimiento & desarrollo , Espermátides/metabolismo , Espermatocitos/citología , Espermatocitos/crecimiento & desarrollo , Espermatocitos/metabolismo , Espermatogonias/citología , Espermatogonias/crecimiento & desarrollo , Espermatogonias/inmunología , Espermatozoides/citología , Espermatozoides/crecimiento & desarrollo , Testículo/citología , Testículo/crecimiento & desarrollo , Testículo/metabolismo
13.
Proc Natl Acad Sci U S A ; 114(47): 12536-12541, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29114052

RESUMEN

Meiotic synapsis and recombination between homologs permits the formation of cross-overs that are essential for generating chromosomally balanced sperm and eggs. In mammals, surveillance mechanisms eliminate meiotic cells with defective synapsis, thereby minimizing transmission of aneuploidy. One such surveillance mechanism is meiotic silencing, the inactivation of genes located on asynapsed chromosomes, via ATR-dependent serine-139 phosphorylation of histone H2AFX (γH2AFX). Stimulation of ATR activity requires direct interaction with an ATR activation domain (AAD)-containing partner. However, which partner facilitates the meiotic silencing properties of ATR is unknown. Focusing on the best-characterized example of meiotic silencing, meiotic sex chromosome inactivation, we reveal this AAD-containing partner to be the DNA damage and checkpoint protein TOPBP1. Conditional TOPBP1 deletion during pachynema causes germ cell elimination associated with defective X chromosome gene silencing and sex chromosome condensation. TOPBP1 is essential for localization to the X chromosome of silencing "sensors," including BRCA1, and effectors, including ATR, γH2AFX, and canonical repressive histone marks. We present evidence that persistent DNA double-strand breaks act as silencing initiation sites. Our study identifies TOPBP1 as a critical factor in meiotic sex chromosome silencing.


Asunto(s)
Proteínas Portadoras/genética , Roturas del ADN de Doble Cadena , Cromosomas Sexuales/química , Espermatogénesis/genética , Inactivación del Cromosoma X , Animales , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteína BRCA1 , Proteínas Portadoras/metabolismo , Emparejamiento Cromosómico , Histonas/genética , Histonas/metabolismo , Masculino , Ratones , Ratones Noqueados , Cromosomas Sexuales/metabolismo , Espermátides/citología , Espermátides/crecimiento & desarrollo , Espermátides/metabolismo , Espermatocitos/citología , Espermatocitos/crecimiento & desarrollo , Espermatocitos/metabolismo , Espermatogonias/citología , Espermatogonias/crecimiento & desarrollo , Espermatogonias/metabolismo , Espermatozoides/citología , Espermatozoides/crecimiento & desarrollo , Espermatozoides/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
14.
J Exp Zool B Mol Dev Evol ; 332(7): 258-268, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31531931

RESUMEN

The DAZ family genes, including boule, dazl, and daz, play pivotal roles in germ cell development and differentiation during gametogenesis in organisms, which have been widely studied in mammals, reptiles, or fishes. Dazl was bisexual expressed in both mitotic and meiotic germ cells, daz was male premeiotic expressed, whereas boule exhibits largely in unisexual meiotic germ cells but bisexual expression in several fishes, however, there is lack of report on boule gene and the evolutionary conservation and divergence of dazl and boule in reptile. Here, both boule and dazl genes were characterized in Pelodiscus sinensis. The quantitative real-time polymerase chain reaction analysis showed that boule and dazl were abundantly expressed in adult ovary and testis but barely in somatic tissues, such as heart, brain, liver, spleen, and kidney. Moreover, through fluorescent in situ hybridization, bisexual and germline-specific expression profiles of boule and dazl messenger RNAs (mRNAs) were demonstrated. Boule mRNA exhibited a maximal meiotic expression in spermatocytes, and a relatively low, but distinct expression in oocytes at meiotic stages in P. sinensis, similar to the expression profile of human boule in ovary. However, dazl mRNA was richly distributed in male germ cells at almost all stages during spermatogenesis, and predominantly expressed in most of stages of oocytes including premeiotic and meiotic stages. These findings imply that boule and dazl would play distinct roles in the sexual differentiation of germ cells during turtle gametogenesis, and the major functions of daz family members involved in germ cell differentiation would be conserved across species including P. sinensis.


Asunto(s)
Proteínas de Unión al ARN/genética , Tortugas/genética , Tortugas/metabolismo , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica , Masculino , Meiosis , Oocitos/crecimiento & desarrollo , Oocitos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Reptiles/genética , Espermatocitos/crecimiento & desarrollo , Espermatocitos/metabolismo
15.
Exp Cell Res ; 371(1): 262-268, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30130520

RESUMEN

The fruit-fly Drosophila melanogaster harbours different types of ciliary structures: ciliary projections associated with neurons of type I and cilium-like regions (CLRs) found during male gametogenesis. The latter deserve particular attention since they are morphologically similar to vertebrate primary cilia and transform into the sperm axonemes during spermiogenesis. Although, all the centrioles are able to organize the CLRs, we found that the mother centriole docks first to the plasma membrane suggesting a new intrinsic functional asymmetry between the parent centrioles. We also show that the CLRs lack the Y-links that connect the axoneme doublets with the plasma membrane in conventional primary cilia. Moreover, the C-tubules, that are lacking in the axoneme of the primary cilia, persisted along the CLRs albeit modified into longitudinal blades. Remarkably, mutant flies in which the CLRs are devoid of the C-tubules or their number is reduced lack sperm axonemes or have incomplete axonemes. Therefore, the C-tubules are dispensable for the assembly of the CLRs but are essential for sperm axoneme elongation and maintenance in Drosophila.


Asunto(s)
Centriolos/ultraestructura , Cilios/ultraestructura , Drosophila melanogaster/ultraestructura , Pupa/ultraestructura , Espermatocitos/ultraestructura , Espermatogénesis/genética , Animales , Axonema/metabolismo , Axonema/ultraestructura , Proteínas de Ciclo Celular/deficiencia , Proteínas de Ciclo Celular/genética , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Centriolos/metabolismo , Cilios/metabolismo , Proteínas de Drosophila/deficiencia , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/metabolismo , Regulación del Desarrollo de la Expresión Génica , Masculino , Meiosis , Microscopía Electrónica de Transmisión , Mutación , Pupa/genética , Pupa/crecimiento & desarrollo , Pupa/metabolismo , Espermatocitos/crecimiento & desarrollo , Espermatocitos/metabolismo
16.
PLoS Genet ; 12(10): e1006389, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27792785

RESUMEN

Mammalian meiocytes feature four meiosis-specific cohesin proteins in addition to ubiquitous ones, but the roles of the individual cohesin complexes are incompletely understood. To decipher the functions of the two meiosis-specific kleisins, REC8 or RAD21L, together with the only meiosis-specific SMC protein SMC1ß, we generated Smc1ß-/-Rec8-/- and Smc1ß-/-Rad21L-/- mouse mutants. Analysis of spermatocyte chromosomes revealed that besides SMC1ß complexes, SMC1α/RAD21 and to a small extent SMC1α/REC8 contribute to chromosome axis length. Removal of SMC1ß and RAD21L almost completely abolishes all chromosome axes. The sex chromosomes do not pair in single or double mutants, and autosomal synapsis is impaired in all mutants. Super resolution microscopy revealed synapsis-associated SYCP1 aberrantly deposited between sister chromatids and on single chromatids in Smc1ß-/-Rad21L-/- cells. All mutants show telomere length reduction and structural disruptions, while wild-type telomeres feature a circular TRF2 structure reminiscent of t-loops. There is no loss of centromeric cohesion in both double mutants at leptonema/early zygonema, indicating that, at least in the mutant backgrounds, an SMC1α/RAD21 complex provides centromeric cohesion at this early stage. Thus, in early prophase I the most prominent roles of the meiosis-specific cohesins are in axis-related features such as axis length, synapsis and telomere integrity rather than centromeric cohesion.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteínas Cromosómicas no Histona/genética , Meiosis/genética , Proteínas Nucleares/genética , Fosfoproteínas/genética , Espermatogénesis/genética , Animales , Centrómero/genética , Cromátides/genética , Emparejamiento Cromosómico/genética , Segregación Cromosómica/genética , Proteínas de Unión al ADN , Masculino , Mamíferos , Profase Meiótica I/genética , Ratones , Ratones Noqueados , Espermatocitos/crecimiento & desarrollo , Complejo Sinaptonémico/genética , Cohesinas
17.
PLoS Genet ; 12(5): e1005863, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27149259

RESUMEN

During transcription, most eukaryotic genes generate multiple alternative cleavage and polyadenylation (APA) sites, leading to the production of transcript isoforms with variable lengths in the 3' untranslated region (3'UTR). In contrast to somatic cells, male germ cells, especially pachytene spermatocytes and round spermatids, express a distinct reservoir of mRNAs with shorter 3'UTRs that are essential for spermatogenesis and male fertility. However, the mechanisms underlying the enrichment of shorter 3'UTR transcripts in the developing male germ cells remain unknown. Here, we report that UPF2-mediated nonsense-mediated mRNA decay (NMD) plays an essential role in male germ cells by eliminating ubiquitous genes-derived, longer 3'UTR transcripts, and that this role is independent of its canonical role in degrading "premature termination codon" (PTC)-containing transcripts in somatic cell lineages. This report provides physiological evidence supporting a noncanonical role of the NMD pathway in achieving global 3'UTR shortening in the male germ cells during spermatogenesis.


Asunto(s)
Proteínas Portadoras/genética , Degradación de ARNm Mediada por Codón sin Sentido/genética , Poliadenilación/genética , Espermatogénesis/genética , Regiones no Traducidas 3'/genética , Animales , Linaje de la Célula/genética , Células Germinativas/crecimiento & desarrollo , Células Germinativas/metabolismo , Masculino , Ratones , ARN Mensajero/genética , Proteínas de Unión al ARN , Transducción de Señal , Espermátides/crecimiento & desarrollo , Espermátides/metabolismo , Espermatocitos/crecimiento & desarrollo , Espermatocitos/metabolismo , Transcripción Genética
18.
PLoS Genet ; 12(1): e1005813, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26808625

RESUMEN

Piwi proteins associate with piRNAs and functions in epigenetic programming, post-transcriptional regulation, transposon silencing, and germline development. However, it is not known whether the diverse functions of these proteins are molecularly separable. Here we report that Piwi interacts with Tudor-SN (Tudor staphylococcal nuclease, TSN) antagonistically in regulating spermatogenesis but synergistically in silencing transposons. However, it is not required for piRNA biogenesis. TSN is known to participate in diverse molecular functions such as RNAi, degradation of hyper-edited miRNAs, and spliceosome assembly. We show that TSN colocalizes with Piwi in primordial germ cells (PGCs) and embryonic somatic cells. In adult ovaries and testes, TSN is ubiquitously expressed and enriched in the cytoplasm of both germline and somatic cells. The tsn mutants display a higher mitotic index of spermatogonia, accumulation of spermatocytes, defects in meiotic cytokinesis, a decreased number of spermatids, and eventually reduced male fertility. Germline-specific TSN-expression analysis demonstrates that this function is germline-dependent. Different from other known Piwi interters, TSN represses Piwi expression at both protein and mRNA levels. Furthermore, reducing piwi expression in the germline rescues tsn mutant phenotype in a dosage-dependent manner, demonstrating that Piwi and TSN interact antagonistically in germ cells to regulate spermatogenesis. However, the tsn deficiency has little, if any, impact on piRNA biogenesis but displays a synergistic effect with piwi mutants in transposon de-silencing. Our results reveal the biological function of TSN and its contrasting modes of interaction with Piwi in spermatogenesis, transposon silencing, and piRNA biogenesis.


Asunto(s)
Proteínas de Drosophila/genética , Epigénesis Genética , Proteínas de Transporte de Membrana/genética , ARN Interferente Pequeño/genética , Espermatogénesis/genética , Animales , Citoplasma/genética , Elementos Transponibles de ADN/genética , Proteínas de Drosophila/biosíntesis , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Masculino , Proteínas de Transporte de Membrana/biosíntesis , Ovario/crecimiento & desarrollo , Ovario/metabolismo , Espermatocitos/crecimiento & desarrollo , Espermatocitos/metabolismo
19.
Biochim Biophys Acta Mol Cell Res ; 1864(3): 546-561, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27993670

RESUMEN

Importin 13 (Imp13) is a bidirectional nuclear transporter of proteins involved in a range of important cellular processes, with an N-terminally truncated inhibitory isoform (tImp13) specifically expressed in testis. To gain insight into tImp13 function, we performed a yeast-2-hybrid screen from a human testis cDNA library, identifying for the first time a suite of interactors with roles in diverse cellular process. We validated the interaction of tImp13 with Eukaryotic translation initiation factor 4γ2 (EIF4G2) and High mobility group containing protein 20A (HMG20A), benchmarking that with glucocorticoid receptor (GR), a known Imp13 interactor expressed in testis. Coimmunoprecipitation assays indicated association of both tImp13 and Imp13 with EIF4G2, HMG20A and GR. Quantitative confocal microscopic analysis revealed the ability of tImp13 to inhibit the nuclear localisation of EIF4G2, HMG20A and GR, as well as that of Imp13 to act as a nuclear exporter for both EIF4G2 and HMG20A, and as a nuclear importer for GR. The physiological relevance of these results was highlighted by the cytoplasmic localisation of EIF4G2, HMG20A and GR in pachytene spermatocytes/round spermatids in the murine testis where tImp13 is present at high levels, in contrast to the nuclear localisation of HMG20A and GR in spermatogonia, where tImp13 is largely absent. Interestingly, Imp13, EIF4G2, HMG20A and GR were found together in the acrosome vesicle of murine epididymal spermatozoa. Collectively, our findings show, for the first time, that tImp13 may have a functional role in the mature spermatozoa, in addition to that in the meiotic germ cells of the testis.


Asunto(s)
Núcleo Celular/metabolismo , Regulación del Desarrollo de la Expresión Génica , Carioferinas/metabolismo , Espermátides/metabolismo , Espermatocitos/metabolismo , Espermatogénesis/genética , Animales , Factor 4G Eucariótico de Iniciación/genética , Factor 4G Eucariótico de Iniciación/metabolismo , Biblioteca de Genes , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/metabolismo , Humanos , Carioferinas/genética , Masculino , Ratones , Unión Proteica , Mapeo de Interacción de Proteínas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transporte de Proteínas , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Transducción de Señal , Espermátides/crecimiento & desarrollo , Espermátides/ultraestructura , Espermatocitos/crecimiento & desarrollo , Espermatocitos/ultraestructura , Testículo/citología , Testículo/crecimiento & desarrollo , Testículo/metabolismo , Técnicas del Sistema de Dos Híbridos
20.
Biochim Biophys Acta Mol Cell Res ; 1864(3): 527-545, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27974247

RESUMEN

Throughout spermatogenesis, two important processes occur at late stage VIII of the seminiferous epithelial cycle in the rat testis: preleptotene spermatocytes commence entry into the adluminal compartment and step 19 spermatids release from the seminiferous epithelium. Presently, it is not clear how these processes, which involve extensive restructuring of unique Sertoli-Sertoli and Sertoli-germ cell junctions, are mediated. We aimed to determine whether annexin A2 (ANXA2), a Ca2+-dependent and phospholipid-binding protein, participates in cell junction dynamics. To address this, in vitro and in vivo RNA interference studies were performed on prepubertal Sertoli cells and adult rat testes. The endpoints of Anxa2 knockdown were determined by immunoblotting, morphological analyses, fluorescent immunostaining, and barrier integrity assays. In the testis, ANXA2 localized to the Sertoli cell stalk, with specific staining at the blood-testis barrier and the concave (ventral) surface of elongated spermatids. ANXA2 also bound actin when testis lysates were used for immunoprecipitation. Anxa2 knockdown was found to disrupt the Sertoli cell/blood-testis barrier in vitro and in vivo. The disruption in barrier function was substantiated by changes in the localization of claudin-11, zona occludens-1, N-cadherin, and ß-catenin. Furthermore, Anxa2 knockdown resulted in spermiation defects caused by a dysfunction of tubulobulbar complexes, testis-specific actin-rich ultrastructures that internalize remnant cell junction components prior to spermiation. Additionally, there were changes in the localization of several tubulobulbar complex component proteins, including actin-related protein 3, cortactin, and dynamin I/II. Our results indicate that ANXA2 is critical for the integrity of the blood-testis barrier and the timely release of spermatids.


Asunto(s)
Anexina A2/genética , Barrera Hematotesticular/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células de Sertoli/metabolismo , Espermátides/metabolismo , Espermatogénesis/genética , Proteína 3 Relacionada con la Actina/genética , Proteína 3 Relacionada con la Actina/metabolismo , Animales , Anexina A2/antagonistas & inhibidores , Anexina A2/metabolismo , Barrera Hematotesticular/crecimiento & desarrollo , Cadherinas/genética , Cadherinas/metabolismo , Claudinas/genética , Claudinas/metabolismo , Cortactina/genética , Cortactina/metabolismo , Dinamina I/genética , Dinamina I/metabolismo , Dinamina II/genética , Dinamina II/metabolismo , Uniones Intercelulares/genética , Masculino , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Epitelio Seminífero/citología , Epitelio Seminífero/crecimiento & desarrollo , Epitelio Seminífero/metabolismo , Células de Sertoli/citología , Transducción de Señal , Espermátides/crecimiento & desarrollo , Espermátides/ultraestructura , Espermatocitos/crecimiento & desarrollo , Espermatocitos/metabolismo , Espermatocitos/ultraestructura , Proteína de la Zonula Occludens-1/genética , Proteína de la Zonula Occludens-1/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA