Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 555
Filtrar
1.
J Dairy Sci ; 106(6): 4366-4379, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37059660

RESUMEN

Misregulation of spermatogenesis transcription factors (TF) in hybrids can lead to misexpression, which is a mechanism for hybrid male sterility (HMS). We used dzo (male offspring of Bos taurus ♂ × Bos grunniens ♀) in bovines to investigate the relationship of the key TF with HMS via RNA sequencing and assay for transposase-accessible chromatin with high-throughput sequencing analyses. RNA sequencing showed that the widespread misexpression in dzo was associated with spermatogenesis-related genes and somatic or progenitor genes. The transition from leptotene or zygotene spermatocytes to pachytene spermatocytes may be the key stage for meiosis arrest in dzo. The analysis of TF-binding motif enrichment revealed that the male meiosis-specific master TF MYB proto-oncogene like 1 (MYBL1, known as A-MYB) motif was enriched on the promoters of downregulated pachytene spermatocyte genes in dzo. Assay for transposase-accessible chromatin with high-throughput sequencing revealed that TF-binding sites for MYBL1, nuclear transcription factor Y, and regulatory factor X were enriched in the low-chromatin accessibility region of dzo. The target genes of the MYBL1-binding motif were associated with meiosis-specific genes and significantly downregulated in dzo testis. The transcription factor MYBL1 may be the candidate master regulator for pachytene spermatocyte genes dysregulated in interspecific HMS dzo. This study reported that a few upstream TF regulation changes might exert a cascading effect downstream in a regulatory network as a mechanism for HMS.


Asunto(s)
Espermatocitos , Factores de Transcripción , Bovinos , Masculino , Animales , Espermatocitos/fisiología , Factores de Transcripción/genética , Espermatogénesis , Testículo , Cromatina
2.
Reprod Domest Anim ; 57(5): 459-464, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35098603

RESUMEN

In mammals, spermatogenesis is a complex and cyclic process in which a spermatogonia turns into a highly differentiated cell: the spermatozoa. Spermatogenesis comprises proliferation of spermatogonia (spermatocytogenesis), meiosis of spermatocytes and finally differentiation of spermatids into spermatozoa (spermiogenesis). This review summarizes the current knowledge on domestic cat spermatogenesis including its physiology, development, efficiency and pathologies as well as their novel non-invasive diagnostic methods. This information will provide a resource for further studies to achieve precise fundamental knowledge of key aspects that will facilitate breeding, management and contraception in this popular species.


Asunto(s)
Espermátides , Espermatogénesis , Animales , Gatos , Masculino , Mamíferos , Meiosis , Espermátides/fisiología , Espermatocitos/fisiología , Espermatogénesis/fisiología , Espermatogonias , Espermatozoides/fisiología , Testículo
3.
Genes (Basel) ; 12(12)2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34946793

RESUMEN

Regulation of transcriptional activity during meiosis depends on the interrelated processes of recombination and synapsis. In eutherian mammal spermatocytes, transcription levels change during prophase-I, being low at the onset of meiosis but highly increased from pachytene up to the end of diplotene. However, X and Y chromosomes, which usually present unsynapsed regions throughout prophase-I in male meiosis, undergo a specific pattern of transcriptional inactivation. The interdependence of synapsis and transcription has mainly been studied in mammals, basically in mouse, but our knowledge in other unrelated phylogenetically species is more limited. To gain new insights on this issue, here we analyzed the relationship between synapsis and transcription in spermatocytes of the grasshopper Eyprepocnemis plorans. Autosomal chromosomes of this species achieve complete synapsis; however, the single X sex chromosome remains always unsynapsed and behaves as a univalent. We studied transcription in meiosis by immunolabeling with RNA polymerase II phosphorylated at serine 2 and found that whereas autosomes are active from leptotene up to diakinesis, the X chromosome is inactive throughout meiosis. This inactivation is accompanied by the accumulation of, at least, two repressive epigenetic modifications: H3 methylated at lysine 9 and H2AX phosphorylated at serine 139. Furthermore, we identified that X chromosome inactivation occurs in premeiotic spermatogonia. Overall, our results indicate: (i) transcription regulation in E. plorans spermatogenesis differs from the canonical pattern found in mammals and (ii) X chromosome inactivation is likely preceded by a process of heterochromatinization before the initiation of meiosis.


Asunto(s)
Saltamontes/genética , Espermatogénesis/genética , Inactivación del Cromosoma X/genética , Cromosoma X/genética , Animales , Emparejamiento Cromosómico/genética , Epigénesis Genética/genética , Femenino , Silenciador del Gen/fisiología , Histonas/genética , Lisina/genética , Masculino , Meiosis/genética , Profase Meiótica I/genética , ARN Polimerasa II/genética , Espermatocitos/fisiología , Cromosoma Y/genética
4.
Front Endocrinol (Lausanne) ; 12: 761249, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34721307

RESUMEN

To achieve spermatogenesis in vitro, one of the most challenging processes to mimic is meiosis. Meiotic problems, like incomplete synapsis of the homologous chromosomes, or impaired homologous recombination, can cause failure of crossover formation and subsequent chromosome nondisjunction, eventually leading to aneuploid sperm. These meiotic events are therefore strictly monitored by meiotic checkpoints that initiate apoptosis of aberrant spermatocytes and lead to spermatogenic arrest. However, we recently found that, in vitro derived meiotic cells proceeded to the first meiotic division (MI) stage, despite displaying incomplete chromosome synapsis, no discernible XY-body and lack of crossover formation. We therefore optimized our in vitro culture system of meiosis from male germline stem cells (mGSCs) in order to achieve full chromosome synapsis, XY-body formation and meiotic crossovers. In comparison to previous culture system, the in vitro-generated spermatocytes were transferred after meiotic initiation to a second culture dish. This dish already contained a freshly plated monolayer of proliferatively inactivated immortalized Sertoli cells supporting undifferentiated mGSCs. In this way we aimed to simulate the multiple layers of germ cell types that support spermatogenesis in vivo in the testis. We found that in this optimized culture system, although independent of the undifferentiated mGSCs, meiotic chromosome synapsis was complete and XY body appeared normal. However, meiotic recombination still occurred insufficiently and only few meiotic crossovers were formed, leading to MI-spermatocytes displaying univalent chromosomes (paired sister chromatids). Therefore, considering that meiotic checkpoints are not necessarily fully functional in vitro, meiotic crossover formation should be closely monitored when mimicking gametogenesis in vitro to prevent generation of aneuploid gametes.


Asunto(s)
Emparejamiento Cromosómico/fisiología , Cromosomas/fisiología , Meiosis/fisiología , Aneuploidia , Animales , Azoospermia/congénito , Azoospermia/fisiopatología , Diferenciación Celular/fisiología , Línea Celular , Proliferación Celular/fisiología , Masculino , Ratones , Ratones Endogámicos DBA , Células de Sertoli/fisiología , Espermatocitos/fisiología , Espermatogénesis/fisiología , Espermatozoides/fisiología , Testículo/fisiología
5.
PLoS Genet ; 17(10): e1009870, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34669718

RESUMEN

Reduction of genome ploidy from diploid to haploid necessitates stable pairing of homologous chromosomes into bivalents before the start of the first meiotic division. Importantly, this chromosome pairing must avoid interlocking of non-homologous chromosomes. In spermatocytes of Drosophila melanogaster, where homolog pairing does not involve synaptonemal complex formation and crossovers, associations between non-homologous chromosomes are broken up by chromosome territory formation in early spermatocytes. Extensive non-homologous associations arise from the coalescence of the large blocks of pericentromeric heterochromatin into a chromocenter and from centromere clustering. Nevertheless, during territory formation, bivalents are moved apart into spatially separate subnuclear regions. The condensin II subunits, Cap-D3 and Cap-H2, have been implicated, but the remarkable separation of bivalents during interphase might require more than just condensin II. For further characterization of this process, we have applied time-lapse imaging using fluorescent markers of centromeres, telomeres and DNA satellites in pericentromeric heterochromatin. We describe the dynamics of the disruption of centromere clusters and the chromocenter in normal spermatocytes. Mutations in Cap-D3 and Cap-H2 abolish chromocenter disruption, resulting in excessive chromosome missegregation during M I. Chromocenter persistence in the mutants is not mediated by the special system, which conjoins homologs in compensation for the absence of crossovers in Drosophila spermatocytes. However, overexpression of Cap-H2 precluded conjunction between autosomal homologs, resulting in random segregation of univalents. Interestingly, Cap-D3 and Cap-H2 mutant spermatocytes displayed conspicuous stretching of the chromocenter, as well as occasional chromocenter disruption, suggesting that territory formation might involve forces unrelated to condensin II. While the molecular basis of these forces remains to be clarified, they are not destroyed by inhibitors of F actin and microtubules. Our results indicate that condensin II activity promotes chromosome territory formation in co-operation with additional force generators and that careful co-ordination with alternative homolog conjunction is crucial.


Asunto(s)
Adenosina Trifosfatasas/genética , Cromosomas/genética , Proteínas de Unión al ADN/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Complejos Multiproteicos/genética , Espermatocitos/fisiología , Animales , Centrómero/genética , Cromatina/genética , Emparejamiento Cromosómico/genética , Segregación Cromosómica/genética , Drosophila melanogaster/metabolismo , Femenino , Heterocromatina/genética , Interfase/genética , Masculino
6.
Mol Biol Rep ; 48(11): 7379-7385, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34626314

RESUMEN

BACKGROUND: Cryptochrome 1 (cry1), the core regulator of the circadian clock, is essential for ontogeny and mammalian reproduction. Unlike in other tissues, the cry1 gene have noncircadian functions in spermatogenesis, which implies the unique role of cry1 gene in the development of testis. The role of cry1 during the puberty has not been described yet. This study aimed to explore the relationship between cry1 expression and spermatogenic cell numbers. METHODS AND RESULTS: We analyzed testicular tissues from Hu sheep aged 0-180 days by hematoxylin and eosin staining, measured cry1 and cell proliferation regulatory factors (bricd5, tnfrsf21, cdk1) expression by quantitative real-time PCR and characterized the transcription factor in the 5' flanking region of cry1 gene. The data revealed that the number of spermatocytes and early spermatocytes increased rapidly from 90 to 120 dpp (day postpartum). Correspondingly, there was a marked variation in the cry1 and cell proliferation related genes (bricd5, tnfrsf21, cdk1) mRNA expression in the testes from the age of 90 days to 180 days (p < 0.05). We also identified some transcription factors (tcfl5) related to cell proliferation. CONCLUSIONS: There is a significant causal relationship between the transcription level of cry1 gene in Hu sheep testes and the number of spermatogenic cells. It is speculated that cry1 gene may regulate the proliferation of spermatogenic cells by regulating the expression of cell proliferation related genes such as bricd5, tnfrsf21 and cdk1.


Asunto(s)
Proteína Quinasa CDC2/genética , Criptocromos/genética , Receptores del Factor de Necrosis Tumoral/genética , Maduración Sexual , Espermatogénesis , Testículo/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proliferación Celular , Regulación del Desarrollo de la Expresión Génica , Masculino , Ovinos , Espermatocitos/metabolismo , Espermatocitos/fisiología , Testículo/crecimiento & desarrollo , Testículo/fisiología
7.
Cells ; 10(9)2021 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-34571960

RESUMEN

Meiosis involves a series of specific chromosome events, namely homologous synapsis, recombination, and segregation. Disruption of either recombination or synapsis in mammals results in the interruption of meiosis progression during the first meiotic prophase. This is usually accompanied by a defective transcriptional inactivation of the X and Y chromosomes, which triggers a meiosis breakdown in many mutant models. However, epigenetic changes and transcriptional regulation are also expected to affect autosomes. In this work, we studied the dynamics of epigenetic markers related to chromatin silencing, transcriptional regulation, and meiotic sex chromosome inactivation throughout meiosis in knockout mice for genes encoding for recombination proteins SPO11, DMC1, HOP2 and MLH1, and the synaptonemal complex proteins SYCP1 and SYCP3. These models are defective in recombination and/or synapsis and promote apoptosis at different stages of progression. Our results indicate that impairment of recombination and synapsis alter the dynamics and localization pattern of epigenetic marks, as well as the transcriptional regulation of both autosomes and sex chromosomes throughout prophase-I progression. We also observed that the morphological progression of spermatocytes throughout meiosis and the dynamics of epigenetic marks are processes that can be desynchronized upon synapsis or recombination alteration. Moreover, we detected an overlap of early and late epigenetic signatures in most mutants, indicating that the normal epigenetic transitions are disrupted. This can alter the transcriptional shift that occurs in spermatocytes in mid prophase-I and suggest that the epigenetic regulation of sex chromosomes, but also of autosomes, is an important factor in the impairment of meiosis progression in mammals.


Asunto(s)
Emparejamiento Cromosómico/genética , Epigénesis Genética/genética , Mamíferos/genética , Meiosis/genética , Proteínas Recombinantes/genética , Recombinación Genética/genética , Animales , Apoptosis/genética , Marcadores Genéticos/genética , Masculino , Ratones , Cromosomas Sexuales/genética , Espermatocitos/fisiología , Transcripción Genética/genética
8.
PLoS Genet ; 17(8): e1009753, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34388164

RESUMEN

Meiosis is essential for the generation of gametes and sexual reproduction, yet the factors and underlying mechanisms regulating meiotic progression remain largely unknown. Here, we showed that MTL5 translocates into nuclei of spermatocytes during zygotene-pachytene transition and ensures meiosis advances beyond pachytene stage. MTL5 shows strong interactions with MuvB core complex components, a well-known transcriptional complex regulating mitotic progression, and the zygotene-pachytene transition of MTL5 is mediated by its direct interaction with the component LIN9, through MTL5 C-terminal 443-475 residues. Male Mtl5c-mu/c-mu mice expressing the truncated MTL5 (p.Ser445Arg fs*3) that lacks the interaction with LIN9 and is detained in cytoplasm showed male infertility and spermatogenic arrest at pachytene stage, same as that of Mtl5 knockout mice, indicating that the interaction with LIN9 is essential for the nuclear translocation and function of MTL5 during meiosis. Our data demonstrated MTL5 translocates into nuclei during the zygotene-pachytene transition to initiate its function along with the MuvB core complex in pachytene spermatocytes, highlighting a new mechanism regulating the progression of male meiosis.


Asunto(s)
Meiosis/fisiología , Metalotioneína/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Animales , Proteínas de Ciclo Celular/metabolismo , Emparejamiento Cromosómico/genética , Citoplasma , Proteínas de Unión al ADN , Fertilidad/genética , Fertilidad/fisiología , Infertilidad Masculina/genética , Infertilidad Masculina/metabolismo , Masculino , Profase Meiótica I/fisiología , Metalotioneína/genética , Ratones , Ratones Endogámicos C57BL , Fase Paquiteno/genética , Espermatocitos/fisiología , Espermatogénesis/fisiología , Testículo , Proteínas Supresoras de Tumor/fisiología
9.
Int J Mol Sci ; 22(16)2021 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-34445597

RESUMEN

Spermatogenesis is a complicated process involving mitotically proliferating spermatogonial cells, meiotically dividing spermatocytes, and spermatid going through maturation into spermatozoa. The post-translational modifications of proteins play important roles in this biological process. S-palmitoylation is one type of protein modifications catalyzed by zinc finger Asp-His-His-Cys (ZDHHC)-family palmitoyl S-acyltransferases. There are 23 mammalian ZDHHCs that have been identified in mouse. Among them, Zdhhc19 is highly expressed in adult testis. However, the in vivo function of Zdhhc19 in mouse spermatogenesis and fertility remains unknown. In this study, we knocked out the Zdhhc19 gene by generating a 2609 bp deletion from exon 3 to exon 6 in mice. No differences were found in testis morphology and testis/body weight ratios upon Zdhhc19 deletion. Spermatogenesis was not disrupted in Zdhhc19 knockout mice, in which properly developed TRA98+ germ cells, SYCP3+ spermatocytes, and TNP1+ spermatids/spermatozoa were detected in seminiferous tubules. Nevertheless, Zdhhc19 knockout mice were male infertile. Zdhhc19 deficient spermatozoa exhibited multiple defects including abnormal morphology of sperm tails and heads, decreased motility, and disturbed acrosome reaction. All of these led to the inability of Zdhhc19 mutant sperm to fertilize oocytes in IVF assays. Taken together, our results support the fact that Zdhhc19 is a testis enriched gene dispensable for spermatogenesis, but is essential for sperm functions in mice.


Asunto(s)
Aciltransferasas/fisiología , Fertilización , Motilidad Espermática , Espermatocitos/citología , Espermatogénesis , Espermatozoides/fisiología , Reacción Acrosómica , Animales , Femenino , Masculino , Ratones , Ratones Noqueados , Espermatocitos/fisiología
10.
Cell Death Dis ; 12(6): 531, 2021 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-34031364

RESUMEN

Monopolar spindle 1 (MPS1), which plays a critical role in somatic mitosis, has also been revealed to be essential for meiosis I in oocytes. Spermatogenesis is an important process involving successive mitosis and meiosis, but the function of MPS1 in spermatogenesis remains unclear. Here, we generated Mps1 conditional knockout mice and found that Ddx4-cre-driven loss of Mps1 in male mice resulted in depletion of undifferentiated spermatogonial cells and subsequently of differentiated spermatogonia and spermatocytes. In addition, Stra8-cre-driven ablation of Mps1 in male mice led to germ cell loss and fertility reduction. Spermatocytes lacking Mps1 have blocked at the zygotene-to-pachytene transition in the prophase of meiosis I, which may be due to decreased H2B ubiquitination level mediated by MDM2. And the expression of many meiotic genes was decreased, while that of apoptotic genes was increased. Moreover, we also detected increased apoptosis in spermatocytes with Mps1 knockout, which may have been the reason why germ cells were lost. Taken together, our findings indicate that MPS1 is required for mitosis of gonocytes and spermatogonia, differentiation of undifferentiated spermatogonia, and progression of meiosis I in spermatocytes.


Asunto(s)
Fertilidad/genética , Proteínas Serina-Treonina Quinasas/fisiología , Espermatogénesis/genética , Animales , Infertilidad Masculina/genética , Masculino , Meiosis/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitosis/genética , Proteínas Serina-Treonina Quinasas/genética , Espermatocitos/fisiología , Espermatogonias/fisiología
11.
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
12.
Elife ; 102021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33973520

RESUMEN

The male germ cells must adopt the correct morphology at each differentiation stage for proper spermatogenesis. The spermatogonia regulates its differentiation state by its own migration. The male germ cells differentiate and mature with the formation of syncytia, failure of forming the appropriate syncytia results in the arrest at the spermatocyte stage. However, the detailed molecular mechanisms of male germ cell morphological regulation are unknown. Here, we found that EXOC1, a member of the Exocyst complex, is important for the pseudopod formation of spermatogonia and spermatocyte syncytia in mice. EXOC1 contributes to the pseudopod formation of spermatogonia by inactivating the Rho family small GTPase Rac1 and also functions in the spermatocyte syncytia with the SNARE proteins STX2 and SNAP23. Since EXOC1 is known to bind to several cell morphogenesis factors, this study is expected to be the starting point for the discovery of many morphological regulators of male germ cells.


Asunto(s)
Espermatocitos/fisiología , Espermatogénesis/genética , Espermatogonias/fisiología , Proteínas de Transporte Vesicular/genética , Animales , Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica , Células Gigantes , Masculino , Ratones , Ratones Endogámicos C57BL , Espermatogonias/citología , Proteínas de Transporte Vesicular/metabolismo
13.
Genes (Basel) ; 12(5)2021 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-33925685

RESUMEN

Nuclear architecture undergoes an extensive remodeling during spermatogenesis, especially at levels of spermatocytes (SPC) and spermatids (SPT). Interestingly, typical events of spermiogenesis, such as nuclear elongation, acrosome biogenesis, and flagellum formation, need a functional cooperation between proteins of the nuclear envelope and acroplaxome/manchette structures. In addition, nuclear envelope plays a key role in chromosome distribution. In this scenario, special attention has been focused on the LINC (linker of nucleoskeleton and cytoskeleton) complex, a nuclear envelope-bridge structure involved in the connection of the nucleoskeleton to the cytoskeleton, governing mechanotransduction. It includes two integral proteins: KASH- and SUN-domain proteins, on the outer (ONM) and inner (INM) nuclear membrane, respectively. The LINC complex is involved in several functions fundamental to the correct development of sperm cells such as head formation and head to tail connection, and, therefore, it seems to be important in determining male fertility. This review provides a global overview of the main LINC complex components, with a special attention to their subcellular localization in sperm cells, their roles in the regulation of sperm morphological maturation, and, lastly, LINC complex alterations associated to male infertility.


Asunto(s)
Núcleo Celular/fisiología , Citoesqueleto/metabolismo , Citoesqueleto/fisiología , Membrana Nuclear/metabolismo , Matriz Nuclear/metabolismo , Espermatozoides/metabolismo , Espermatozoides/fisiología , Animales , Núcleo Celular/metabolismo , Humanos , Infertilidad Masculina/metabolismo , Infertilidad Masculina/fisiopatología , Masculino , Mecanotransducción Celular/fisiología , Matriz Nuclear/fisiología , Espermátides/metabolismo , Espermátides/fisiología , Espermatocitos/metabolismo , Espermatocitos/fisiología
14.
Am J Hum Genet ; 108(2): 324-336, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-33508233

RESUMEN

Human infertility is a multifactorial disease that affects 8%-12% of reproductive-aged couples worldwide. However, the genetic causes of human infertility are still poorly understood. Synaptonemal complex (SC) is a conserved tripartite structure that holds homologous chromosomes together and plays an indispensable role in the meiotic progression. Here, we identified three homozygous mutations in the SC coding gene C14orf39/SIX6OS1 in infertile individuals from different ethnic populations by whole-exome sequencing (WES). These mutations include a frameshift mutation (c.204_205del [p.His68Glnfs∗2]) from a consanguineous Pakistani family with two males suffering from non-obstructive azoospermia (NOA) and one female diagnosed with premature ovarian insufficiency (POI) as well as a nonsense mutation (c.958G>T [p.Glu320∗]) and a splicing mutation (c.1180-3C>G) in two unrelated Chinese men (individual P3907 and individual P6032, respectively) with meiotic arrest. Mutations in C14orf39 resulted in truncated proteins that retained SYCE1 binding but exhibited impaired polycomplex formation between C14ORF39 and SYCE1. Further cytological analyses of meiosis in germ cells revealed that the affected familial males with the C14orf39 frameshift mutation displayed complete asynapsis between homologous chromosomes, while the affected Chinese men carrying the nonsense or splicing mutation showed incomplete synapsis. The phenotypes of NOA and POI in affected individuals were well recapitulated by Six6os1 mutant mice carrying an analogous mutation. Collectively, our findings in humans and mice highlight the conserved role of C14ORF39/SIX6OS1 in SC assembly and indicate that the homozygous mutations in C14orf39/SIX6OS1 described here are responsible for infertility of these affected individuals, thus expanding our understanding of the genetic basis of human infertility.


Asunto(s)
Azoospermia/genética , Mutación , Insuficiencia Ovárica Primaria/genética , Adulto , Azoospermia/fisiopatología , Emparejamiento Cromosómico , Codón sin Sentido , Proteínas de Unión al ADN/metabolismo , Femenino , Homocigoto , Humanos , Masculino , Meiosis , Persona de Mediana Edad , Proteínas Nucleares/metabolismo , Linaje , Insuficiencia Ovárica Primaria/fisiopatología , Espermatocitos/metabolismo , Espermatocitos/fisiología , Complejo Sinaptonémico/genética , Complejo Sinaptonémico/metabolismo , Secuenciación Completa del Genoma
15.
Front Endocrinol (Lausanne) ; 12: 807374, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35046903

RESUMEN

Fine particulate matter (PM2.5)-induced male reproductive toxicity arouses global public health concerns. However, the mechanisms of toxicity remain unclear. This study aimed to further investigate toxicity pathways by exposure to PM2.5in vitro and in vivo through the application of metabolomics and transcriptomics. In vitro, spermatocyte-derived GC-2spd cells were treated with 0, 25, 50, 100 µg/mL PM2.5 for 48 h. In vivo, the real-world exposure of PM2.5 for mouse was established. Forty-five male C57BL/6 mice were exposed to filtered air, unfiltered air, and concentrated ambient PM2.5 in Tangshan of China for 8 weeks, respectively. The results in vitro and in vivo showed that PM2.5 exposure inhibited GC-2spd cell proliferation and reduced sperm motility. Mitochondrial damage was observed after PM2.5 treatment. Increased Humanin and MOTS-c levels and decreased mitochondrial respiratory indicated that mitochondrial function was disturbed. Furthermore, nontargeted metabolomics analysis revealed that PM2.5 exposure could disturb the citrate cycle (TCA cycle) and reduce amino acids and nucleotide synthesis. Mechanically, the aryl hydrocarbon receptor (AhR) pathway was activated after exposure to PM2.5, with a significant increase in CYP1A1 expression. Further studies showed that PM2.5 exposure significantly increased both intracellular and mitochondrial reactive oxygen species (ROS) and activated NRF2 antioxidative pathway. With the RNA-sequencing technique, the differentially expressed genes induced by PM2.5 exposure were mainly enriched in the metabolism of xenobiotics by the cytochrome P450 pathway, of which Cyp1a1 was the most significantly changed gene. Our findings demonstrated that PM2.5 exposure could induce spermatocyte damage and energy metabolism disorder. The activation of the aryl hydrocarbon receptor might be involved in the mechanism of male reproductive toxicity.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Metabolismo Energético , Material Particulado/toxicidad , Receptores de Hidrocarburo de Aril/metabolismo , Reproducción , Espermatocitos/fisiología , Animales , Línea Celular , Ciclo del Ácido Cítrico , Sistema Enzimático del Citocromo P-450/metabolismo , Perfilación de la Expresión Génica , Masculino , Metaboloma , Metabolómica , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Motilidad Espermática , Transcriptoma , Xenobióticos/metabolismo
16.
Andrology ; 9(2): 665-672, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33089633

RESUMEN

BACKGROUND: MicroRNAs play a crucial role in the regulation of spermatogenesis. For example, miR-128-3p expression is known to decrease significantly after testicular hyperthermia, but the regulatory effect of this change on the spermatogenesis damage caused by heat stress remains unclear. OBJECTIVES: This study aimed to verify whether the target gene of miR-128-3p is MAPK14, which affects spermatogenic cell proliferation and apoptosis under testicular hyperthermia. MATERIALS AND METHODS: Mouse testis and GC2 spermatocyte cell line heat stress models were established. miR-128-3p expression before and after heat stress was analyzed by reverse transcription polymerase chain reaction. MAPK14 and p-MAPK14 expression was detected by Western blot, and cell apoptosis was analyzed by Annexin V-FITC/PI. Subsequently, miR-128-3p inhibitors and mimics were used to interfere with spermatocytes before and after heat stress, respectively, for correlation detection. RESULTS: Compared with the control group, the heat stress group showed decreased miR-128-3p expression, increased p-MAPK14 expression, and decreased cell proliferation activity. In the GC2-spd cell line in vitro, miR-128-3p inhibitors were found to upregulate p-MAPK14 expression, reduce cell proliferation activity, and increase apoptosis, consistent with the results obtained in the heat treatment alone. Furthermore, miR-128-3p mimics transfected in the GC2 cells after heat stress reduced p-MAPK14 expression, alleviated the decrease in cell proliferation, and decreased the apoptosis level. CONCLUSIONS: The downregulation of miR-128-3p expression plays an important role in spermatogenesis damages after testicular hyperthermia, which is probably attributable to the activation of the MAPK signaling pathway. Downregulated miR-128-3p expression induces the apoptosis and inhibits the proliferation of spermatogenic cells by promoting MAPK14 phosphorylation.


Asunto(s)
Apoptosis/genética , MicroARNs/fisiología , Espermatocitos/fisiología , Testículo/metabolismo , Animales , Línea Celular , Activación Enzimática/genética , Regulación del Desarrollo de la Expresión Génica , Respuesta al Choque Térmico , Masculino , Ratones , Ratones Endogámicos ICR , Proteína Quinasa 14 Activada por Mitógenos , Espermatocitos/enzimología , Espermatogénesis/genética , Testículo/citología , Testículo/enzimología
17.
Gene ; 764: 145080, 2021 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-32858178

RESUMEN

Spermatocyte (spc) formation from spermatogonia (spg) differentiation is the first step of spermatogenesis which produces prodigious spermatozoa for a lifetime. After decades of studies, several factors involved in the functioning of a mouse were discovered both inside and outside spg. Considering the peculiar expression and working pattern of each factor, this review divides the whole conversion of spg to spc into four consecutive development processes with a focus on extracellular cues and downstream transcription network in each one. Potential coordination among Dmrt1, Sohlh1/2 and BMP families mediates Ngn3 upregulation, which marks progenitor spg, with other changes. After that, retinoic acid (RA), as a master regulator, promotes A1 spg formation with its helpers and Sall4. A1-to-B spg transition is under the control of Kitl and impulsive RA signaling together with early and late transcription factors Stra8 and Dmrt6. Finally, RA and its responsive effectors conduct the entry into meiosis. The systematic transcription network from outside to inside still needs research to supplement or settle the controversials in each process. As a step further ahead, this review provides possible drug targets for infertility therapy by cross-linking humans and mouse model.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Espermatocitos/fisiología , Espermatogénesis/genética , Espermatogonias/fisiología , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Autorrenovación de las Células/genética , Humanos , Masculino , Ratones , Túbulos Seminíferos/citología , Túbulos Seminíferos/crecimiento & desarrollo , Factores de Transcripción/metabolismo , Transcripción Genética , Tretinoina/metabolismo , Regulación hacia Arriba
18.
Reprod Fertil Dev ; 32(18): 1357-1364, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33308394

RESUMEN

In this study, the ability of cold-induced RNA-binding protein (CIRBP) to regulate the expression of Src-associated during mitosis of 68 kDa (Sam68) and extracellular signal-regulated kinases (ERK) in the mouse testis and mouse primary spermatocytes (GC-2spd cell line) before and after heat stress was examined to explore the molecular mechanism by which CIRBP decreases testicular injury. A mouse testicular hyperthermia model, a mouse primary spermatocyte hyperthermia model and a low CIRBP gene-expression cell model were constructed and their relevant parameters were analysed. The mRNA and protein levels of CIRBP and Sam68 were significantly decreased in the 3-h and 12-h testicular heat-stress groups, extracellular signal-regulated kinase 1/2 (ERK1/2) protein expression was not significantly affected but phospho-ERK1/2 protein levels were significantly decreased. GC-2spd cellular heat-stress results showed that the mRNA and protein concentrations of CIRBP and Sam68 were reduced 48h after heat stress. In the low CIRBP gene-expression cell model, CIRBP protein expression was significantly decreased. Sam68 mRNA expression was significantly decreased only at the maximum transfection concentration of 50nM and Sam68 protein expression was not significantly affected. These findings suggest that CIRBP may regulate the expression of Sam68 at the transcriptional level and the expression of phospho-ERK1/2 protein, both of which protect against heat-stress-induced testicular injury in mice.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Quinasas MAP Reguladas por Señal Extracelular/genética , Respuesta al Choque Térmico/fisiología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/fisiología , Enfermedades Testiculares , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Apoptosis/genética , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación de la Expresión Génica , Respuesta al Choque Térmico/genética , Masculino , Ratones , Ratones Endogámicos ICR , Proteínas de Unión al ARN/metabolismo , Espermatocitos/patología , Espermatocitos/fisiología , Enfermedades Testiculares/etiología , Enfermedades Testiculares/genética , Enfermedades Testiculares/metabolismo , Enfermedades Testiculares/patología , Testículo/metabolismo , Testículo/patología
19.
Mol Reprod Dev ; 87(6): 680-691, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32506506

RESUMEN

Humans are occasionally exposed to extreme environmental heat for a prolonged period of time. Here, we investigated testicular responses to whole-body heat exposure by placing mice in a warm chamber. Among the examined tissues, the testis was found to be most susceptible to heat stress. Heat stress induces direct responses within germ cells, such as eukaryotic initiation factor 2α phosphorylation and stress granule (SG) formation. Prolonged heat stress (42°C for 6 hr) also disturbed tissue organization, such as through blood-testis barrier (BTB) leakage. Germ cell apoptosis was induced by heat stress for 6 hr in a cell type- and developmental stage-specific manner. We previously showed that spermatocytes in the early tubular stages (I-VI) form SGs for protection against heat stress. In the mid-tubular stages (VII-VIII), BTB leakage synergistically enhances the adverse effects of heat stress on pachytene spermatocyte apoptosis. In the late tubular stages (IX-XII), SGs are not formed and severe leakage of the BTB does not occur, resulting in mild apoptosis of late-pachytene spermatocytes near meiosis. Our results revealed that multiple stress responses are involved in germ cell damage resulting from prolonged heat stress (42°C for 6 hr).


Asunto(s)
Apoptosis/fisiología , Respuesta al Choque Térmico/fisiología , Espermatogénesis/fisiología , Espermatozoides/fisiología , Animales , Diferenciación Celular/fisiología , Calor , Masculino , Ratones , Espermatocitos/citología , Espermatocitos/fisiología , Testículo/citología , Testículo/fisiología , Factores de Tiempo
20.
Reprod Domest Anim ; 55(8): 998-1010, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32515513

RESUMEN

The region-specific expression of seminal proteins in testis and excurrent duct system determines the quality and function of the spermatozoa. In the present study, localization and expression of some of the seminal proteins such as insulin-like growth factor receptor 1ß (IGF-1Rß), phosphatidylethanolamine-binding protein 4 (PEBP4), α-tubulin and tissue factor pathway inhibitor 2 (TFPI2) were carried out in testis, excurrent duct system and spermatozoa of buffalo. IGF-1Rß was localized in the cells of the seminiferous tubules of the testis, except in primary spermatocytes. The PEBP4 was localized only in the elongated spermatid, whereas α-tubulin and TFPI2 proteins were localized in all cells of the seminiferous tubule including spermatocyte. In the buffalo spermatozoa, IGF-1Rß, PEBP4, α-tubulin and TFPI2 were localized in the acrosome region, the post-acrosomal region till the tail end, post-acrosome to the entire tail region and the equatorial region, respectively. The study indicates that IGF-1R, α-tubulin and PEBP4 proteins regulate spermatogenesis, whereas TFPI2 may be involved during the zona binding process of the buffalo spermatozoa.


Asunto(s)
Búfalos/fisiología , Proteínas de Plasma Seminal/metabolismo , Espermatogénesis/fisiología , Testículo/metabolismo , Animales , Masculino , Análisis de Semen , Túbulos Seminíferos , Espermatocitos/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...