Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
Cell Commun Signal ; 22(1): 267, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38745232

RESUMO

Low sperm motility is a significant contributor to male infertility. beta-defensins have been implicated in host defence and the acquisition of sperm motility; however, the regulatory mechanisms governing their gene expression patterns and functions remain poorly understood. In this study, we performed single-cell RNA and spatial transcriptome sequencing to investigate the cellular composition of testicular and epididymal tissues and examined their gene expression characteristics. In the epididymis, we found that epididymal epithelial cells display a region specificity of gene expression in different epididymal segments, including the beta-defensin family genes. In particular, Defb15, Defb18, Defb20, Defb25 and Defb48 are specific to the caput; Defb22, Defb23 and Defb26 to the corpus; Defb2 and Defb9 to the cauda of the epididymis. To confirm this, we performed mRNA fluorescence in situ hybridisation (FISH) targeting certain exon region of beta-defensin genes, and found some of their expression matched the sequencing results and displayed a close connection with epididimosome marker gene Cd63. In addition, we paid attention to the Sertoli cells and Leydig cells in the testis, along with fibroblasts and smooth muscle cells in the epididymis, by demonstrating their gene expression profile and spatial information. Our study provides a single-cell and spatial landscape for analysing the gene expression characteristics of testicular and epididymal environments and has important implications for the study of spermatogenesis and sperm maturation.


Assuntos
Epididimo , Análise de Célula Única , Maturação do Esperma , Transcriptoma , beta-Defensinas , Masculino , Animais , beta-Defensinas/genética , beta-Defensinas/metabolismo , Camundongos , Transcriptoma/genética , Maturação do Esperma/genética , Epididimo/metabolismo , Espermatozoides/metabolismo , Família Multigênica , Camundongos Endogâmicos C57BL , Testículo/metabolismo
2.
BMC Bioinformatics ; 25(1): 29, 2024 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-38233783

RESUMO

The impairment of sperm maturation is one of the major pathogenic factors in male subfertility, a serious medical and social problem affecting millions of global couples. Regrettably, the existing research on sperm maturation is slow, limited, and fragmented, largely attributable to the lack of a global molecular view. To fill the data gap, we newly established a database, namely the Sperm Maturation Database (SperMD, http://bio-add.org/SperMD ). SperMD integrates heterogeneous multi-omics data (170 transcriptomes, 91 proteomes, and five human metabolomes) to illustrate the transcriptional, translational, and metabolic manifestations during the entire lifespan of sperm maturation. These data involve almost all crucial scenarios related to sperm maturation, including the tissue components of the epididymal microenvironment, cell constituents of tissues, different pathological states, and so on. To the best of our knowledge, SperMD could be one of the limited repositories that provide focused and comprehensive information on sperm maturation. Easy-to-use web services are also implemented to enhance the experience of data retrieval and molecular comparison between humans and mice. Furthermore, the manuscript illustrates an example application demonstrated to systematically characterize novel gene functions in sperm maturation. Nevertheless, SperMD undertakes the endeavor to integrate the islanding omics data, offering a panoramic molecular view of how the spermatozoa gain full reproductive abilities. It will serve as a valuable resource for the systematic exploration of sperm maturation and for prioritizing the biomarkers and targets for precise diagnosis and therapy of male subfertility.


Assuntos
Infertilidade Masculina , Maturação do Esperma , Masculino , Humanos , Animais , Camundongos , Maturação do Esperma/genética , Sêmen , Espermatozoides/metabolismo , Epididimo/metabolismo , Infertilidade Masculina/metabolismo
3.
Zhonghua Nan Ke Xue ; 29(2): 99-105, 2023 Feb.
Artigo em Chinês | MEDLINE | ID: mdl-37847080

RESUMO

OBJECTIVE: To investigate whether mouse epididymis-specific mRNAs Adam7 and Crisp1 can be delivered into N2a and TM4 cells, and to provide an experimental basis for exploring the function of epididymal mRNAs. METHODS: Using RT-PCR, we detected the presence of epididymis-specific genes (Adam7, Crisp1, Defb22, Wfdc2, and Wfdc9) in the testis, epididymis, epididymosome and sperm of adult male BALB/c mice as well as in the human testis, seminal vesicles and sperm. We isolated epididymosomes of BALB/c mice by low-speed centrifugation, filtration and ultracentrifugation, fluorescently labeled them by PKH26, co-incubated them for 1 hour with the N2a and TM4 cells after 24 hours of starvation culture, and observed whether they were fused with the N2a and TM4 cells and ingested using the epididymosomes without PKH26 labeling, PKH26 dye without epididymosomes, and non- epididymosome or -PKH26 dye as controls. Then we detected the epididymis-specific genes in the N2a and TM4 cells after 1-hour co-incubation by RT-PCR. RESULTS: Adam7 and Crisp1 were present in the mouse epididymis, epididymosomes and sperm, and in the human seminal vesicles and sperm as well, but not in the testes of either the mice or men. PKH26 and Hoechst33258 fluorescence double-labeling showed that the mouse epididymosomes were fused with the N2a and TM4 cells and ingested; RT-PCR revealed the mRNAs of Adam7 and Crisp1 in the N2a and TM4 cells after 1-hour co-incubation; and Western blot exhibited the CRISP1 protein in the N2a and TM4 cells incubated with epididymosomes. CONCLUSION: Epididymosomes can deliver epididymis-specific mRNAs Adam7 and Crisp1 into N2a and TM4 cells, where Crisp1 may be translated into proteins, though their function and significance need to be further studied.


Assuntos
Epididimo , Testículo , Masculino , Humanos , Camundongos , Animais , Testículo/metabolismo , Maturação do Esperma/genética , Sêmen , Espermatozoides/metabolismo , Proteína 2 do Domínio Central WAP de Quatro Dissulfetos/metabolismo
4.
Dev Cell ; 58(9): 779-790.e4, 2023 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-37023748

RESUMO

Reports that mouse sperm gain small RNAs from the epididymosomes secreted by epididymal epithelial cells and that these "foreign" small RNAs act as an epigenetic information carrier mediating the transmission of acquired paternal traits have drawn great attention because the findings suggest that heritable information can flow from soma to germ line, thus invalidating the long-standing Weismann's barrier theory on heritable information flow. Using small RNA sequencing (sRNA-seq), northern blots, sRNA in situ hybridization, and immunofluorescence, we detected substantial changes in the small RNA profile in murine caput epididymal sperm (sperm in the head of the epididymis), and we further determined that the changes resulted from sperm exchanging small RNAs, mainly tsRNAs and rsRNAs, with cytoplasmic droplets rather than the epididymosomes. Moreover, the murine sperm-borne small RNAs were mainly derived from the nuclear small RNAs in late spermatids. Thus, caution is needed regarding sperm gaining foreign small RNAs as an underlying mechanism of epigenetic inheritance.


Assuntos
Epididimo , MicroRNAs , Masculino , Camundongos , Animais , Maturação do Esperma/genética , Sêmen , Espermatozoides , MicroRNAs/genética , Espermátides
5.
Mol Biol (Mosk) ; 56(4): 585-594, 2022.
Artigo em Russo | MEDLINE | ID: mdl-35964315

RESUMO

ALDH4A1, a member of the aldehyde dehydrogenase superfamily, is a key enzyme in the mitochondrial proline metabolism pathway. Recent studies have shown that mutations in aldh4a1 lead to reduced fertility and reproductive premature aging of male nematodes. However, the effect of ALDH4A1 on fertility of male mice has not been studied. In this study, we used CRISPR-Cas9 technology to construct a knockout mouse model of Aldh4a1 for the first time to explore the effect of this gene on the reproduction of male mice. The results showed that compared with WT male mice, Aldh4a1^(-/-) male mice were fertile, had normal spermatogenesis but defect in sperm maturation in the epididymis documented by impaired motility, increased morphological abnormalities and increased spontaneous acrosome reaction. In addition, transmission electron microscopy showed vacuoles in the sperm mitochondria, and fracture in the neck of sperms and vacuoles in these mice. These results revealed that ALDH4A1 plays a vital role in the structure of sperm flagellum and the process of sperm maturation in mice.


Assuntos
Sêmen , Maturação do Esperma , Animais , Masculino , Camundongos , Camundongos Knockout , Maturação do Esperma/genética , Espermatogênese/genética , Espermatozoides
6.
Anim Sci J ; 93(1): e13732, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35543176

RESUMO

The first filial generation of the cattleyaks demonstrates hybrid vigor; however, the male cattleyaks are infertile and restrict productivity and breeding. The discovery of genes in a segment-specific approach offers valuable information and understanding concerning fertility status, yet the biology of cattleyak epididymis is still progressing. Comparative transcriptome analysis was performed on segment pairs of cattleyak epididymis. The caput versus corpus epididymis provided the highest (57.8%) differentially expressed genes (DEGs), corpus versus cauda (25.1%) followed, whereas caput versus cauda pair (17.1%) had the least DEGs. The expression levels of genes coding EPHB6, TLR1, MUC20, MT3, INHBB, TRPV5, EI24, PAOX, KIF12, DEPDC5, and KRT25, which might have the potentials to regulate the homeostasis, innate immunity, differentiation, motility, transport, and sperm maturation-related function in epididymal cells, were downregulated in the distal segment of epididymis. Top enriched KEGG pathways included mTOR, axon guidance, and taste transduction signaling pathways. EIF4B, EPHB6, and TAS2R42 were enriched in the pathways, respectively. Identifying key, new, and unexplored DEGs among the epididymal segments and further analyzing them could boost cattleyak fertility by maximizing sperm quality from genetically better sires and also facilitate better understanding of the epididymal biology.


Assuntos
Epididimo , Maturação do Esperma , Animais , Epididimo/metabolismo , Perfilação da Expressão Gênica/veterinária , Masculino , RNA-Seq/veterinária , Maturação do Esperma/genética , Espermatozoides/metabolismo
7.
Genes (Basel) ; 13(2)2022 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-35205340

RESUMO

The hypothalamus-pituitary-testis axis controls the production of spermatozoa, and the kisspeptin system, comprising Kiss1 and Kiss1 receptor (Kiss1R), is the main central gatekeeper. The activity of the kisspeptin system also occurs in testis and spermatozoa, but currently the need of peripheral kisspeptin to produce gametes is not fully understood. Hence, we characterized kisspeptin system in rat spermatozoa and epididymis caput and cauda and analyzed the possible presence of Kiss1 in the epididymal fluid. The presence of Kiss1 and Kiss1R in spermatozoa collected from epididymis caput and cauda was evaluated by Western blot; significant high Kiss1 levels in the caput (p < 0.001 vs. cauda) and constant levels of Kiss1R proteins were observed. Immunofluorescence analysis revealed that the localization of Kiss1R in sperm head shifts from the posterior region in the epididymis caput to perforatorium in the epididymis cauda. In spermatozoa-free epididymis, Western blot revealed higher expression of Kiss1 and Kiss1R in caput (p < 0.05 vs. cauda). Moreover, immunohistochemistry revealed that Kiss1 and Kiss1R proteins were mainly localized in the secretory epithelial cell types and in contractile myoid cells, respectively. Finally, both dot blot and Elisa revealed the presence of Kiss1 in the epididymal fluid collected from epididymis cauda and caput, indicating that rat epididymis and spermatozoa possess a complete kisspeptin system. In conclusion, we reported for the first time in rodents Kiss1R trafficking in spermatozoa during the epididymis transit and Kiss1 measure in the epididymal fluid, thus suggesting a possible role for the system in spermatozoa maturation and storage within the epididymis.


Assuntos
Epididimo , Kisspeptinas , Animais , Epididimo/metabolismo , Kisspeptinas/genética , Kisspeptinas/metabolismo , Masculino , Proteínas/metabolismo , Ratos , Receptores de Kisspeptina-1/genética , Receptores de Kisspeptina-1/metabolismo , Maturação do Esperma/genética , Espermatozoides/metabolismo
8.
Hum Reprod ; 37(4): 651-668, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35137089

RESUMO

STUDY QUESTION: Are epididymosomes implicated in protein transfer from the epididymis to spermatozoa? SUMMARY ANSWER: We characterized the contribution of epididymal secretions to the sperm proteome and demonstrated that sperm acquire epididymal proteins through epididymosomes. WHAT IS KNOWN ALREADY: Testicular sperm are immature cells unable to fertilize an oocyte. After leaving the testis, sperm transit along the epididymis to acquire motility and fertilizing abilities. It is well known that marked changes in the sperm proteome profile occur during epididymal maturation. Since the sperm is a transcriptional and translational inert cell, previous studies have shown that sperm incorporate proteins, RNA and lipids from extracellular vesicles (EVs), released by epithelial cells lining the male reproductive tract. STUDY DESIGN, SIZE, DURATION: We examined the contribution of the epididymis to the post-testicular maturation of spermatozoa, via the production of EVs named epididymosomes, released by epididymal epithelial cells. An integrative analysis using both human and mouse data was performed to identify sperm proteins with a potential epididymis-derived origin. Testes and epididymides from adult humans (n = 9) and adult mice (n = 3) were used to experimentally validate the tissue localization of four selected proteins using high-resolution confocal microscopy. Mouse epididymal sperm were co-incubated with carboxyfluorescein succinimidyl ester (CFSE)-labeled epididymosomes (n = 4 mice), and visualized using high-resolution confocal microscopy. PARTICIPANTS/MATERIALS, SETTING, METHODS: Adult (12-week-old) C57BL/CBAF1 wild-type male mice and adult humans were used for validation purposes. Testes and epididymides from both mice and humans were obtained and processed for immunofluorescence. Mouse epididymal sperm and mouse epididymosomes were obtained from the epididymal cauda segment. Fluorescent epididymosomes were obtained after labeling the epididymal vesicles with CFSE dye followed by epididymosome isolation using a density cushion. Immunofluorescence was performed following co-incubation of sperm with epididymosomes in vitro. High-resolution confocal microscopy and 3D image reconstruction were used to visualize protein localization and sperm-epididymosomes interactions. MAIN RESULTS AND THE ROLE OF CHANCE: Through in silico analysis, we first identified 25 sperm proteins with a putative epididymal origin that were conserved in both human and mouse spermatozoa. From those, the epididymal origin of four sperm proteins (SLC27A2, EDDM3B, KRT19 and WFDC8) was validated by high-resolution confocal microscopy. SLC27A2, EDDM3B, KRT19 and WFDC8 were all detected in epithelial cells lining the human and mouse epididymis, and absent from human and mouse seminiferous tubules. We found region-specific expression patterns of these proteins throughout the mouse epididymides. In addition, while EDDM3B, KRT19 and WFDC8 were detected in both epididymal principal and clear cells (CCs), SLC27A2 was exclusively expressed in CCs. Finally, we showed that CFSE-fluorescently labeled epididymosomes interact with sperm in vitro and about 12-36% of the epididymosomes contain the targeted sperm proteins with an epididymal origin. LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: The human and mouse sample size was limited and our results were descriptive. The analyses of epididymal sperm and epididymosomes were solely performed in the mouse model due to the difficulties in obtaining epididymal luminal fluid human samples. Alternatively, human ejaculated sperm and seminal EVs could not be used because ejaculated sperm have already contacted with the fluids secreted by the male accessory sex glands, and seminal EVs contain other EVs in addition to epididymosomes, such as the abundant prostate-derived EVs. WIDER IMPLICATIONS OF THE FINDINGS: Our findings indicate that epididymosomes are capable of providing spermatozoa with a new set of epididymis-derived proteins that could modulate the sperm proteome and, subsequently, participate in the post-testicular maturation of sperm cells. Additionally, our data provide further evidence of the novel role of epididymal CCs in epididymosome production. Identifying mechanisms by which sperm mature to acquire their fertilization potential would, ultimately, lead to a better understanding of male reproductive health and may help to identify potential therapeutic strategies to improve male infertility. STUDY FUNDING/COMPETING INTEREST(S): This work was supported by the Spanish Ministry of Economy and Competitiveness (Ministerio de Economía y Competividad; fondos FEDER 'una manera de hacer Europa' PI13/00699 and PI16/00346 to R.O.; and Sara Borrell Postdoctoral Fellowship, Acción Estratégica en Salud, CD17/00109 to J.C.), by National Institutes of Health (grants HD040793 and HD069623 to S.B., grant HD104672-01 to M.A.B.), by the Spanish Ministry of Education, Culture and Sports (Ministerio de Educación, Cultura y Deporte para la Formación de Profesorado Universitario, FPU15/02306 to F.B.), by a Lalor Foundation Fellowship (to F.B. and M.A.B.), by the Government of Catalonia (Generalitat de Catalunya, pla estratègic de recerca i innovació en salut, PERIS 2016-2020, SLT002/16/00337 to M.J.), by Fundació Universitària Agustí Pedro i Pons (to F.B.), and by the American Society for Biochemistry and Molecular Biology (PROLAB Award from ASBMB/IUBMB/PABMB to F.B.). Confocal microscopy and transmission electron microscopy was performed in the Microscopy Core facility of the Massachusetts General Hospital (MGH) Center for Systems Biology/Program in Membrane Biology which receives support from Boston Area Diabetes and Endocrinology Research Center (BADERC) award DK57521 and Center for the Study of Inflammatory Bowel Disease grant DK43351. The Zeiss LSM800 microscope was acquired using an NIH Shared Instrumentation Grant S10-OD-021577-01. The authors have no conflicts of interest to declare.


Assuntos
Epididimo , Maturação do Esperma , Animais , Epididimo/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Maturação do Esperma/genética , Espermatozoides/metabolismo , Testículo
9.
PLoS Genet ; 17(3): e1009416, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33661909

RESUMO

Beyond the haploid genome, mammalian sperm carry a payload of epigenetic information with the potential to modulate offspring phenotypes. Recent studies show that the small RNA repertoire of sperm is remodeled during post-testicular maturation in the epididymis. Epididymal maturation has also been linked to changes in the sperm methylome, suggesting that the epididymis might play a broader role in shaping the sperm epigenome. Here, we characterize the genome-wide methylation landscape in seven germ cell populations from throughout the male reproductive tract. We find very few changes in the cytosine methylation landscape between testicular germ cell populations and cauda epididymal sperm, demonstrating that the sperm methylome is stable throughout post-testicular maturation. Although our sequencing data suggested that caput epididymal sperm exhibit a highly unusual methylome, follow-up studies revealed that this resulted from contamination of caput sperm by extracellular DNA. Extracellular DNA formed web-like structures that ensnared sperm, and was present only in sperm samples obtained from the caput epididymis and vas deferens of virgin males. Curiously, contaminating extracellular DNA was associated with citrullinated histone H3, potentially resulting from a PAD-driven genome decondensation process. Taken together, our data emphasize the stability of cytosine methylation in mammalian sperm, and identify a surprising, albeit transient, period during which sperm are associated with extracellular DNA.


Assuntos
Citosina/metabolismo , Metilação de DNA , Epigenoma , Maturação do Esperma/genética , Espermatozoides/metabolismo , Testículo/metabolismo , Animais , Diferenciação Celular/genética , Ácidos Nucleicos Livres , Ilhas de CpG , Epididimo/citologia , Epididimo/metabolismo , Feminino , Masculino , Camundongos , Espermatozoides/citologia
10.
Reprod Domest Anim ; 56(4): 555-576, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33438262

RESUMO

The epididymis is the site of post-testicular sperm maturation, which constitutes the acquisition of sperm motility and the ability to recognize and fertilize oocytes. The role of miRNA in male reproductive system, including the control of different steps leading to proper fertilization such as gametogenesis, sperm maturation and maintenance of male fertility where the deletion of Dicer in mouse germ cells led to infertility, has been demonstrated. The identification of miRNA expression in a region-specific manner will therefore provide valuable insight into the functional differences between the regions of the epididymis. In this study, we employed RNA-seq technology to explore the expression pattern of miRNAs and establish some miRNAs of significant interest with regard to epididymal sperm maturation in the CY epididymis. We identified a total of 431 DE known miRNAs; 119, 185 and 127 DE miRNAs were detected for caput versus corpus, corpus versus cauda and caput versus cauda region pairs, respectively. Our results demonstrate region-specific miRNA expression in the CY epididymis. The GO and KEGG enrichment for the predicted target genes indicated the functional values of miRNAs. Furthermore, we observed that the expression of miR-200a was downregulated in the caput, compared with cauda. Since the family of miR-200 has previously been suggested to contribute to the distinct physiological function of sperm maturation in epididymis of adult rat, we speculate that the downregulation of miR-200a in CY caput epididymis may play an important role of sperm maturation in the epididymis of CY. Therefore, our findings may not only increase our understanding of the molecular mechanisms regulated by the miRNA functions in region-specific miRNA expression in the CY epididymis, it could provide a valuable information to understand the mechanism of male infertility of CY.


Assuntos
Bovinos/fisiologia , Epididimo/metabolismo , MicroRNAs/metabolismo , Maturação do Esperma/fisiologia , Animais , Epididimo/anatomia & histologia , Hibridização Genética/genética , Infertilidade Masculina/genética , Infertilidade Masculina/veterinária , Masculino , MicroRNAs/genética , RNA-Seq/veterinária , Maturação do Esperma/genética
11.
Poult Sci ; 99(11): 6094-6118, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33142529

RESUMO

The application of transcriptomics to the study of the reproductive tract in male turkeys can significantly increase our current knowledge regarding the specifics of bird reproduction. To characterize the complex transcriptomic changes that occur in the testis, epididymis, and ductus deferens, deep sequencing of male turkey RNA samples (n = 6) was performed, using Illumina RNA-Seq. The obtained sequence reads were mapped to the turkey genome, and relative expression values were calculated to analyze differentially expressed genes (DEGs). Statistical analysis revealed 1,682; 2,150; and 340 DEGs in testis/epididymis, testis/ductus deferens, and epididymis/ductus deferens comparisons, respectively. The expression of selected genes was validated using quantitative real-time reverse transcriptase-polymerase chain reaction. Bioinformatics analysis revealed several potential candidate genes involved in spermatogenesis, spermiogenesis and flagellum formation in the testis, and in post-testicular sperm maturation in the epididymis and ductus deferens. In the testis, genes were linked with the mitotic proliferation of spermatogonia and the meiotic division of spermatocytes. Histone ubiquitination and protamine phosphorylation were shown to be regulatory mechanisms for nuclear condensation during spermiogenesis. The characterization of testicular transcripts allowed a better understanding of acrosome formation and development and flagellum formation, including axoneme structures and functions. Spermatozoa motility during post-testicular maturation was linked to the development of flagellar actin filaments and biochemical processes, including Ca2+ influx and protein phosphorylation/dephosphorylation. Spermatozoa quality appeared to be controlled by apoptosis and antioxidant systems in the epididymis and ductus deferens. Finally, genes associated with reproductive system development and morphogenesis were identified. To the best of our knowledge, this is the first genome-wide functional investigation of genes associated with tissue-specific processes in turkey reproductive tract. A catalog of genes worthy of further studies to understand the avian reproductive physiology and regulation was provided.


Assuntos
Genitália Masculina , Maturação do Esperma , Espermatogênese , Transcriptoma , Perus , Animais , Perfilação da Expressão Gênica/veterinária , Genitália Masculina/metabolismo , Masculino , Maturação do Esperma/genética , Espermatogênese/genética , Espermatozoides , Testículo , Perus/genética , Perus/crescimento & desenvolvimento , Perus/metabolismo
12.
Dev Dyn ; 249(12): 1500-1513, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32959928

RESUMO

BACKGROUND: The transcription factor pleomorphic adenoma gene 1 (PLAG1) is required for male fertility. Mice deficient in PLAG1 exhibit decreased sperm motility and abnormal epididymal tubule elongation and coiling, indicating impaired sperm maturation during epididymal transit. However, the downstream transcriptomic profile of the Plag1 knockout (KO; Plag1-/- ) murine epididymis is currently unknown. RESULTS: In this study, the PLAG1-dependent epididymal transcriptome was characterised using RNA sequencing. Several genes important for the control of sperm maturation, motility, capacitation and the acrosome reaction were dysregulated in Plag1-/- mice. Surprisingly, several cell proliferation genes were upregulated, and Ki67 analysis indicated that cell proliferation is aberrantly upregulated in the cauda epididymis stroma of Plag1-/- mice. Gene ontology analysis showed an overall upregulation of genes encoding extracellular matrix components, and an overall downregulation of genes encoding metalloendopeptidases in the epididymides from Plag1-/- mice. CONCLUSION: Together, these results suggest a defect in the epididymal extracellular matrix in Plag1-/- mice. These results imply that in addition to maintaining epididymal integrity directly, PLAG1 may also regulate several genes involved in the regulation of sperm maturation and capacitation. Moreover, PLAG1 may also be involved in regulating tissue homeostasis and ensuring proper structure and maintenance of the extracellular matrix in the epididymis.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Epididimo/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Maturação do Esperma/genética , Transcriptoma , Animais , Proteínas de Ligação a DNA/genética , Proteínas da Matriz Extracelular/genética , Perfilação da Expressão Gênica , Masculino , Camundongos , Camundongos Knockout
13.
Int J Mol Sci ; 21(15)2020 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-32751076

RESUMO

It is well-established that testicular spermatozoa are immature and acquire motility and fertilization capabilities during transit throughout the epididymis. The epididymis is a duct-like organ that connects the testis to the vas deferens and is comprised of four anatomical regions: the initial segment, caput, corpus, and cauda. Sperm maturation occurs during epididymal transit by the interaction of sperm cells with the unique luminal environment of each epididymal region. In this review we discuss the epididymis as an essential reproductive organ responsible for sperm concentration, maturation (including sperm motility acquisition and fertilizing ability), protection and storage. Importantly, we also discuss specific characteristics and roles of epididymal-derived exosomes (epididymosomes) in establishing sperm competency within the intricate process of reproduction. This review suggests that an increasing body of evidence is working to develop a complete picture of the role of the epididymis in male reproduction, offspring health, and disease susceptibility.


Assuntos
Epididimo/metabolismo , Fertilização/genética , Reprodução/genética , Maturação do Esperma/genética , Espermatozoides/metabolismo , Animais , Epididimo/citologia , Epigênese Genética , Exossomos/genética , Exossomos/metabolismo , Feminino , Humanos , Padrões de Herança , Masculino , Camundongos , Oócitos/citologia , Oócitos/metabolismo , Motilidade dos Espermatozoides/genética , Espermatozoides/citologia , Testículo/citologia , Testículo/metabolismo , Ducto Deferente/citologia , Ducto Deferente/metabolismo
14.
Chromosoma ; 129(3-4): 215-226, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32651609

RESUMO

Heterochromatin protein 1ß (HP1ß), encoded by the Cbx1 gene, has been functionally linked to chromatin condensation, transcriptional regulation, and DNA damage repair. Here we report that testis-specific Cbx1 conditional knockout (Cbx1 cKO) impairs male germ cell development in mice. Depletion of HP1ß negatively affected sperm maturation and increased seminiferous tubule degeneration in Cbx1 cKO mice. In addition, the spermatogonia have elevated γ-H2AX foci levels as do Cbx1 deficient mouse embryonic fibroblasts (MEFs) as compared to wild-type (WT) control MEFs. The increase in γ-H2AX foci in proliferating Cbx1 cKO cells indicates defective replication-dependent DNA damage repair. Depletion or loss of HP1ß from human cells and MEFs increased DNA replication fork stalling and firing of new origins of replication, indicating defective DNA synthesis. Taken together, these results suggest that loss of HP1ß in proliferating cells leads to DNA replication defects with associated DNA damage that impact spermatogenesis.


Assuntos
Proteínas Cromossômicas não Histona/genética , Replicação do DNA , Regulação da Expressão Gênica no Desenvolvimento , Espermatogênese/genética , Alelos , Animais , Apoptose/genética , Linhagem Celular , Homólogo 5 da Proteína Cromobox , Proteínas Cromossômicas não Histona/metabolismo , Replicação do DNA/efeitos dos fármacos , Imunofluorescência , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Marcação de Genes , Loci Gênicos , Histonas/metabolismo , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Knockout , Fenótipo , Maturação do Esperma/genética , Espermatogênese/efeitos dos fármacos , Espermatogônias/citologia , Espermatogônias/metabolismo
15.
Biol Reprod ; 103(3): 521-533, 2020 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-32412041

RESUMO

During spermiogenesis in mammals, actin filaments and a variety of actin-binding proteins are involved in the formation and function of highly specialized testis-specific structures. Actin-based motor proteins, such as myosin Va and VIIa, play a key role in this complex process of spermatid transformation into mature sperm. We have previously demonstrated that myosin VI (MYO6) is also expressed in mouse testes. It is present in actin-rich structures important for spermatid development, including one of the earliest events in spermiogenesis-acrosome formation. Here, we demonstrate using immunofluorescence, cytochemical, and ultrastructural approaches that MYO6 is involved in maintaining the structural integrity of these specialized actin-rich structures during acrosome biogenesis in mouse. We show that MYO6 together with its binding partner TOM1/L2 is present at/around the spermatid Golgi complex and the nascent acrosome. Depletion of MYO6 in Snell's waltzer mice causes structural disruptions of the Golgi complex and affects the acrosomal granule positioning within the developing acrosome. In summary, our results suggest that MYO6 plays an anchoring role during the acrosome biogenesis mainly by tethering of different cargo/membranes to highly specialized actin-related structures.


Assuntos
Acrossomo/metabolismo , Acrossomo/ultraestrutura , Cadeias Pesadas de Miosina/biossíntese , Espermatogênese/fisiologia , Reação Acrossômica , Actinas/metabolismo , Animais , Complexo de Golgi/genética , Complexo de Golgi/metabolismo , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Cadeias Pesadas de Miosina/genética , Contagem de Espermatozoides , Maturação do Esperma/genética , Espermátides
16.
Nat Commun ; 11(1): 1499, 2020 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-32198406

RESUMO

Extracellular vesicles (EVs) are a unique mode of intercellular communication capable of incredible specificity in transmitting signals involved in cellular function, including germ cell maturation. Spermatogenesis occurs in the testes, behind a protective barrier to ensure safeguarding of germline DNA from environmental insults. Following DNA compaction, further sperm maturation occurs in the epididymis. Here, we report reproductive tract EVs transmit information regarding stress in the paternal environment to sperm, potentially altering fetal development. Using intracytoplasmic sperm injection, we found that sperm incubated with EVs collected from stress-treated epididymal epithelial cells produced offspring with altered neurodevelopment and adult stress reactivity. Proteomic and transcriptomic assessment of these EVs showed dramatic changes in protein and miRNA content long after stress treatment had ended, supporting a lasting programmatic change in response to chronic stress. Thus, EVs as a normal process in sperm maturation, can also perform roles in intergenerational transmission of paternal environmental experience.


Assuntos
Vesículas Extracelulares/metabolismo , Sistema Nervoso/crescimento & desenvolvimento , Proteômica , Reprodução/fisiologia , Adolescente , Animais , Técnicas de Cultura de Células , Epididimo/metabolismo , Epigênese Genética , Epigenômica , Feminino , Células Germinativas , Histonas , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/metabolismo , Nanopartículas , Maturação do Esperma/genética , Maturação do Esperma/fisiologia , Espermatogênese/genética , Espermatogênese/fisiologia , Espermatozoides/metabolismo , Estresse Fisiológico , Testículo
17.
Andrologia ; 52(1): e13352, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31746491

RESUMO

Histone phosphorylation, an epigenetic post-translational modification, plays essential roles in male gamete chromatin compaction during spermatogenesis and sperm maturity. Previously, we studied the epigenetic marker of phosphorylated serine 1 of histone H2A and H4 (HS1ph) during spermatogenesis in mice and crabs, which was shown to be closely related to the sperm maturity. To further investigate the correlation between phosphorylated serine 1 of histone H4 (H4S1ph) and sperm maturation, a comparison study was conducted in this work between the healthy and the precocious crabs. It was discovered that the distribution of H4S1ph was similar for the two groups of crabs during spermatogenesis before maturity, but totally different in the sperm nuclei. H4S1ph vanished in the nuclei of healthy crab spermatozoa mostly, while retained in the precocious crabs just like what it was in elongated spermatid of both kinds of crabs. The results showed that a high level of H4S1ph conservation was closely associated with immaturity and might indicate inefficient fertility of male precocious crabs. Thus, H4S1ph was suggested to be an epigenetic marker of sperm maturity.


Assuntos
Epigênese Genética/fisiologia , Histonas/genética , Puberdade Precoce/genética , Maturação do Esperma/genética , Animais , Braquiúros , Núcleo Celular/genética , Núcleo Celular/metabolismo , Modelos Animais de Doenças , Fertilidade/genética , Histonas/metabolismo , Humanos , Masculino , Fosforilação , Serina/metabolismo , Espermatozoides/fisiologia , Testículo/anatomia & histologia , Testículo/fisiologia
18.
Elife ; 82019 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-31687931

RESUMO

Tandemly-repeated DNAs, or satellites, are enriched in heterochromatic regions of eukaryotic genomes and contribute to nuclear structure and function. Some satellites are transcribed, but we lack direct evidence that specific satellite RNAs are required for normal organismal functions. Here, we show satellite RNAs derived from AAGAG tandem repeats are transcribed in many cells throughout Drosophila melanogaster development, enriched in neurons and testes, often localized within heterochromatic regions, and important for viability. Strikingly, we find AAGAG transcripts are necessary for male fertility, and that AAGAG RNA depletion results in defective histone-protamine exchange, sperm maturation and chromatin organization. Since these events happen late in spermatogenesis when the transcripts are not detected, we speculate that AAGAG RNA in primary spermatocytes 'primes' post-meiosis steps for sperm maturation. In addition to demonstrating essential functions for AAGAG RNAs, comparisons between closely related Drosophila species suggest that satellites and their transcription evolve quickly to generate new functions.


Assuntos
Drosophila melanogaster/genética , Fertilidade/genética , Regulação da Expressão Gênica no Desenvolvimento , Repetições de Microssatélites , RNA Mensageiro/genética , Maturação do Esperma/genética , Espermatogênese/genética , Animais , Drosophila melanogaster/citologia , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Embrião não Mamífero , Evolução Molecular , Heterocromatina/metabolismo , Heterocromatina/ultraestrutura , Histonas/metabolismo , Masculino , Protaminas/metabolismo , RNA Mensageiro/biossíntese , Espermatócitos/citologia , Espermatócitos/crescimento & desenvolvimento , Espermatócitos/metabolismo , Transcrição Gênica
19.
Andrology ; 7(5): 741-747, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31050198

RESUMO

BACKGROUND: The epithelial lining of the human epididymis is critical for sperm maturation. This process requires distinct specialized functions in the head, body, and tail of the duct. These region-specific properties are maintained by distinct gene expression profiles which are governed by transcription factor networks, non-coding RNAs, and other factors. MATERIALS AND METHODS: We used genome-wide protocols including DNase-seq, RNA-seq and ChIP-seq to characterize open (active) chromatin, the transcriptome and occupancy of specific transcription factors (TFs) respectively, in caput, corpus, and cauda segments of adult human epididymis tissue and primary human epididymis epithelial (HEE) cell cultures derived from them. RNA-seq following TF depletion or activation, combined with gene ontology analysis also determined TF targets. RESULTS: Among regional differentially expressed transcripts were epithelial-selective transcription factors (TFs), microRNAs, and antiviral response genes. Caput-enriched TFs included hepatocyte nuclear factor 1 (HNF1) and the androgen receptor (AR), both of which were also predicted to occupy cis-regulatory elements identified as open chromatin in HEE cells. HNF1 targets were identified genome-wide using ChIP-seq, in HEE cells. Next, siRNA-mediated depletion of HNF1 revealed a pivotal role for this TF in coordinating epithelial water and solute transport in caput epithelium. The importance of AR in HEE cells was shown by AR ChIP-seq, and by RNA-seq after synthetic androgen (R1881) treatment. AR has a distinct transcriptional program in the HEE cells and likely recruits different co-factors (RUNX1 and CEBPß) in comparison to those used in prostate epithelium. DISCUSSION AND CONCLUSION: Our data identify many transcription factors that regulate the development and differentiation of HEE cells. Moreover, a comparison between immature and adult HEE cells showed key TFs in the transition to fully differentiated function of this epithelium. These data may help identify new targets to treat male infertility and have the potential to open new avenues for male contraception.


Assuntos
Biologia Computacional/métodos , Epididimo/metabolismo , Regulação da Expressão Gênica/genética , Redes Reguladoras de Genes/genética , Perfilação da Expressão Gênica , Fator 1 Nuclear de Hepatócito/metabolismo , Humanos , Masculino , Maturação do Esperma/genética , Transcriptoma/genética
20.
Gene ; 706: 201-210, 2019 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-31085275

RESUMO

The functional sperm is the key factor for species continuation. The process spermatogenesis, to produce mature sperm is quite complex. It begins with the proliferation and differentiation of spermatogonia, which develop from primary spermatocytes to secondary spermatocytes and round spermatids, which eventually develop into fertile mature sperm. Spermiogenesis is the latest stage of spermatogenesis, where the round spermatids undergo a series of dramatic morphological changes and extreme condensation of chromatin to construct mature sperm with species-specific shape. During spermiogenesis, chromatin remodeling is a unique progress. It leads the nucleosome from a histone-based structure to a mostly protamine-based configuration. The main events of chromatin remodeling are the replacement of histone by histone variants, hyperacetylation, transient DNA strand breaks and repair, variants by transition proteins and finally by protamines. In this review, we synthesize and summarize the current knowledge on the progress of chromatin remodeling during spermiogenesis. We straighten out the chronological order of chromatin remodeling and illustrate the possible regulation mechanisms of each step.


Assuntos
Montagem e Desmontagem da Cromatina/fisiologia , Cromatina/fisiologia , Espermatogênese/fisiologia , Animais , Cromatina/genética , Montagem e Desmontagem da Cromatina/genética , DNA/metabolismo , Histonas/metabolismo , Humanos , Masculino , Maturação do Esperma/genética , Espermátides/metabolismo , Espermatócitos/metabolismo , Espermatogênese/genética , Espermatozoides/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA