Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Genome Res ; 31(7): 1174-1186, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34158368

RESUMEN

The testis transcriptome is highly complex and includes RNAs that potentially hybridize to form double-stranded RNA (dsRNA). We isolated dsRNA using the monoclonal J2 antibody and deep-sequenced the enriched samples from testes of juvenile Dicer1 knockout mice, age-matched controls, and adult animals. Comparison of our data set with recently published data from mouse liver revealed that the dsRNA transcriptome in testis is markedly different from liver: In testis, dsRNA-forming transcripts derive from mRNAs including promoters and immediate downstream regions, whereas in somatic cells they originate more often from introns and intergenic transcription. The genes that generate dsRNA are significantly expressed in isolated male germ cells with particular enrichment in pachytene spermatocytes. dsRNA formation is lower on the sex (X and Y) chromosomes. The dsRNA transcriptome is significantly less complex in juvenile mice as compared to adult controls and, possibly as a consequence, the knockout of Dicer1 has only a minor effect on the total number of transcript peaks associated with dsRNA. The comparison between dsRNA-associated genes in testis and liver with a reported set of genes that produce endogenous siRNAs reveals a significant overlap in testis but not in liver. Testis dsRNAs also significantly associate with natural antisense genes-again, this feature is not observed in liver. These findings point to a testis-specific mechanism involving natural antisense transcripts and the formation of dsRNAs that feed into the RNA interference pathway, possibly to mitigate the mutagenic impacts of recombination and transposon mobilization.

2.
Nucleic Acids Res ; 50(20): 11470-11491, 2022 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-36259644

RESUMEN

Nonsense-mediated RNA decay (NMD) is a highly conserved and selective RNA turnover pathway that depends on the endonuclease SMG6. Here, we show that SMG6 is essential for male germ cell differentiation in mice. Germ-cell conditional knockout (cKO) of Smg6 induces extensive transcriptome misregulation, including a failure to eliminate meiotically expressed transcripts in early haploid cells, and accumulation of NMD target mRNAs with long 3' untranslated regions (UTRs). Loss of SMG6 in the male germline results in complete arrest of spermatogenesis at the early haploid cell stage. We find that SMG6 is strikingly enriched in the chromatoid body (CB), a specialized cytoplasmic granule in male germ cells also harboring PIWI-interacting RNAs (piRNAs) and the piRNA-binding protein PIWIL1. This raises the possibility that SMG6 and the piRNA pathway function together, which is supported by several findings, including that Piwil1-KO mice phenocopy Smg6-cKO mice and that SMG6 and PIWIL1 co-regulate many genes in round spermatids. Together, our results demonstrate that SMG6 is an essential regulator of the male germline transcriptome, and highlight the CB as a molecular platform coordinating RNA regulatory pathways to control sperm production and fertility.


Asunto(s)
Endorribonucleasas , Gránulos de Ribonucleoproteína de Células Germinales , Espermatogénesis , Transcriptoma , Animales , Masculino , Ratones , Células Germinativas/metabolismo , ARN Interferente Pequeño/genética , Espermátides/metabolismo , Espermatogénesis/genética , Endorribonucleasas/metabolismo
3.
Reproduction ; 165(1): 49-63, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36194437

RESUMEN

In brief: Proper regulation of heterochromatin is critical for spermatogenesis. This study reveals the dynamic localization patterns of distinct chromatin regulators during spermatogenesis and disrupted sex chromatin status in spermatocytes in the absence of DICER. Abstract: Heterochromatin is dynamically formed and organized in differentiating male germ cells, and its proper regulation is a prerequisite for normal spermatogenesis. While heterochromatin is generally transcriptionally silent, we have previously shown that major satellite repeat (MSR) DNA in the pericentric heterochromatin (PCH) is transcribed during spermatogenesis. We have also shown that DICER associates with PCH and is involved in the regulation of MSR-derived transcripts. To shed light on the heterochromatin regulation in the male germline, we studied the expression, localization and heterochromatin association of selected testis-enriched chromatin regulators in the mouse testis. Our results show that HELLS, WDHD1 and BAZ1A are dynamically expressed during spermatogenesis. They display limited overlap in expression, suggesting involvement in distinct heterochromatin-associated processes at different steps of differentiation. We also show that HELLS and BAZ1A interact with DICER and MSR chromatin. Interestingly, deletion of Dicer1 affects the sex chromosome heterochromatin status in late pachytene spermatocytes, as demonstrated by mislocalization of Polycomb protein family member SCML1 to the sex body. These data substantiate the importance of dynamic heterochromatin regulation during spermatogenesis and emphasize the key role of DICER in the maintenance of chromatin status in meiotic male germ cells.


Asunto(s)
Cromatina , Proteínas Cromosómicas no Histona , ADN Helicasas , Heterocromatina , Animales , Masculino , Ratones , Cromatina/metabolismo , ADN Helicasas/genética , Heterocromatina/metabolismo , Espermatocitos/metabolismo , Espermatogénesis/fisiología , Testículo/metabolismo , Proteínas Cromosómicas no Histona/genética
4.
Genes Dev ; 29(10): 1032-44, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25995188

RESUMEN

MIWI catalytic activity is required for spermatogenesis, indicating that piRNA-guided cleavage is critical for germ cell development. To identify meiotic piRNA targets, we augmented the mouse piRNA repertoire by introducing a human meiotic piRNA cluster. This triggered a spermatogenesis defect by inappropriately targeting the piRNA machinery to mouse mRNAs essential for germ cell development. Analysis of such de novo targets revealed a signature for pachytene piRNA target recognition. This enabled identification of both transposable elements and meiotically expressed protein-coding genes as targets of native piRNAs. Cleavage of genic targets began at the pachytene stage and resulted in progressive repression through meiosis, driven at least in part via the ping-pong cycle. Our data support the idea that meiotic piRNA populations must be strongly selected to enable successful spermatogenesis, both driving the response away from essential genes and directing the pathway toward mRNA targets that are regulated by small RNAs in meiotic cells.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Meiosis , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Espermatogénesis/genética , Animales , Elementos Transponibles de ADN/genética , Silenciador del Gen , Humanos , Infertilidad Masculina/genética , Masculino , Ratones , Sistemas de Lectura Abierta/genética , Fase Paquiteno/genética , Testículo/metabolismo
5.
Nucleic Acids Res ; 48(13): 7135-7153, 2020 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-32484548

RESUMEN

Constitutive heterochromatin at the pericentric regions of chromosomes undergoes dynamic changes in its epigenetic and spatial organization during spermatogenesis. Accurate control of pericentric heterochromatin is required for meiotic cell divisions and production of fertile and epigenetically intact spermatozoa. In this study, we demonstrate that pericentric heterochromatin is expressed during mouse spermatogenesis to produce major satellite repeat (MSR) transcripts. We show that the endonuclease DICER localizes to the pericentric heterochromatin in the testis. Furthermore, DICER forms complexes with MSR transcripts, and their processing into small RNAs is compromised in Dicer1 knockout mice leading to an elevated level of MSR transcripts in meiotic cells. We also show that defective MSR forward transcript processing in Dicer1 cKO germ cells is accompanied with reduced recruitment of SUV39H2 and H3K9me3 to the pericentric heterochromatin and meiotic chromosome missegregation. Altogether, our results indicate that the physiological role of DICER in maintenance of male fertility extends to the regulation of pericentric heterochromatin through direct targeting of MSR transcripts.


Asunto(s)
ARN Helicasas DEAD-box/fisiología , Ribonucleasa III/fisiología , Espermátides , Espermatocitos , Espermatogénesis , Testículo/metabolismo , Animales , Centrómero/metabolismo , Segregación Cromosómica , Fertilidad , Heterocromatina/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Masculino , Meiosis/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Espermátides/citología , Espermátides/metabolismo , Espermatocitos/citología , Espermatocitos/metabolismo , Secuencias Repetidas en Tándem/genética , Testículo/citología
6.
Development ; 144(14): 2683-2693, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28619825

RESUMEN

Sperm differentiation requires specific protein transport for correct sperm tail formation and head shaping. A transient microtubular structure, the manchette, appears around the differentiating spermatid head and serves as a platform for protein transport to the growing tail. Sperm flagellar 2 (SPEF2) is known to be essential for sperm tail development. In this study we investigated the function of SPEF2 during spermatogenesis using a male germ cell-specific Spef2 knockout mouse model. In addition to defects in sperm tail development, we observed a duplication of the basal body and failure in manchette migration resulting in an abnormal head shape. We identified cytoplasmic dynein 1 and GOLGA3 as novel interaction partners for SPEF2. SPEF2 and dynein 1 colocalize in the manchette and the inhibition of dynein 1 disrupts the localization of SPEF2 to the manchette. Furthermore, the transport of a known SPEF2-binding protein, IFT20, from the Golgi complex to the manchette was delayed in the absence of SPEF2. These data indicate a possible novel role of SPEF2 as a linker protein for dynein 1-mediated cargo transport along microtubules.


Asunto(s)
Proteínas/fisiología , Espermátides/crecimiento & desarrollo , Espermátides/fisiología , Espermatogénesis/fisiología , Animales , Proteínas Portadoras/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Dineínas Citoplasmáticas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microtúbulos/fisiología , Transporte de Proteínas/genética , Transporte de Proteínas/fisiología , Proteínas/genética , Cola del Espermatozoide/fisiología , Cola del Espermatozoide/ultraestructura , Espermátides/citología , Espermatogénesis/genética
7.
FASEB J ; 32(6): 3229-3241, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29401623

RESUMEN

The pituitary gonadotrophins and testosterone are the main hormonal regulators of spermatogenesis, but estradiol is also known to play a role in the process. The hormonal responses in the testis are partially mediated by somatic Sertoli cells that provide nutritional and physical support for differentiating male germ cells. Hydroxysteroid (17ß) dehydrogenase 1 (HSD17B1) is a steroidogenic enzyme that especially catalyzes the conversion of low potent 17keto-steroids to highly potent 17ß-hydroxysteroids. In this study, we show that Hsd17b1 is highly expressed in Sertoli cells of fetal and newborn mice, and HSD17B1 knockout males present with disrupted spermatogenesis with major defects, particularly in the head shape of elongating spermatids. The cell-cell junctions between Sertoli cells and germ cells were disrupted in the HSD17B1 knockout mice. This resulted in complications in the orientation of elongating spermatids in the seminiferous epithelium, reduced sperm production, and morphologically abnormal spermatozoa. We also showed that the Sertoli cell-expressed HSD17B1 participates in testicular steroid synthesis, evidenced by a compensatory up-regulation of HSD17B3 in Leydig cells. These results revealed a novel role for HSD17B1 in the control of spermatogenesis and male fertility, and that Sertoli cells significantly contribute to steroid synthesis in the testis.-Hakkarainen, J., Zhang, F.-P., Jokela, H., Mayerhofer, A., Behr, R., Cisneros-Montalvo, S., Nurmio, M., Toppari, J., Ohlsson, C., Kotaja, N., Sipilä, P., Poutanen, M. Hydroxysteroid (17ß) dehydrogenase 1 expressed by Sertoli cells contributes to steroid synthesis and is required for male fertility.


Asunto(s)
17-Hidroxiesteroide Deshidrogenasas/biosíntesis , Fertilidad/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , Células de Sertoli/enzimología , Espermatogénesis/fisiología , Esteroides/biosíntesis , 17-Hidroxiesteroide Deshidrogenasas/genética , Animales , Masculino , Ratones , Ratones Noqueados , Epitelio Seminífero/citología , Epitelio Seminífero/enzimología , Células de Sertoli/citología , Espermátides/citología , Espermátides/enzimología
8.
Reproduction ; 155(2): R77-R91, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29038333

RESUMEN

Germ cells have exceptionally diverse transcriptomes. Furthermore, the progress of spermatogenesis is accompanied by dramatic changes in gene expression patterns, the most drastic of them being near-to-complete transcriptional silencing during the final steps of differentiation. Therefore, accurate RNA regulatory mechanisms are critical for normal spermatogenesis. Cytoplasmic germ cell-specific ribonucleoprotein (RNP) granules, known as germ granules, participate in posttranscriptional regulation in developing male germ cells. Particularly, germ granules provide platforms for the PIWI-interacting RNA (piRNA) pathway and appear to be involved both in piRNA biogenesis and piRNA-targeted RNA degradation. Recently, other RNA regulatory mechanisms, such as the nonsense-mediated mRNA decay pathway have also been associated to germ granules providing new exciting insights into the function of germ granules. In this review article, we will summarize our current knowledge on the role of germ granules in the control of mammalian male germ cell's transcriptome and in the maintenance of fertility.


Asunto(s)
Regulación de la Expresión Génica , Células Germinativas/fisiología , ARN Interferente Pequeño/genética , Proteínas de Unión al ARN/metabolismo , Espermatogénesis , Animales , Células Germinativas/citología , Humanos , Masculino
9.
RNA ; 20(4): 483-95, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24554440

RESUMEN

The genome of male germ cells is actively transcribed during spermatogenesis to produce phase-specific protein-coding mRNAs and a considerable amount of different noncoding RNAs. Ribonucleoprotein (RNP) granule-mediated RNA regulation provides a powerful means to secure the quality and correct expression of the requisite transcripts. Haploid spermatids are characterized by a unique, unusually large cytoplasmic granule, the chromatoid body (CB), which emerges during the switch between the meiotic and post-meiotic phases of spermatogenesis. To better understand the role of the CB in male germ cell differentiation, we isolated CBs from mouse testes and revealed its full RNA and protein composition. We showed that the CB is mainly composed of RNA-binding proteins and other proteins involved RNA regulation. The CB was loaded with RNA, including pachytene piRNAs, a diverse set of mRNAs, and a number of uncharacterized long noncoding transcripts. The CB was demonstrated to accumulate nascent RNA during all the steps of round spermatid differentiation. Our results revealed the CB as a large germ cell-specific RNP platform that is involved in the control of the highly complex transcriptome of haploid male germ cells.


Asunto(s)
Gránulos Citoplasmáticos/fisiología , Células Germinativas/metabolismo , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Espermátides/metabolismo , Espermatogénesis/fisiología , Animales , Biomarcadores/metabolismo , Gránulos Citoplasmáticos/ultraestructura , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Células Germinativas/ultraestructura , Masculino , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espermátides/ultraestructura
10.
Biol Reprod ; 93(6): 139, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26510868

RESUMEN

The endonuclease DICER that processes micro-RNAs and small interfering RNAs is essential for normal spermatogenesis and male fertility. We previously showed that the deletion of Dicer1 gene in postnatal spermatogonia in mice using Ngn3 promoter-driven Cre expression caused severe defects in the morphogenesis of haploid spermatid to mature spermatozoon, including problems in cell polarization and nuclear elongation. In this study, we further analyzed the same mouse model and revealed that absence of functional DICER in differentiating male germ cells induces disorganization of the cell-cell junctions in the seminiferous epithelium. We detected discontinuous and irregular apical ectoplasmic specializations between elongating spermatids and Sertoli cells. The defective anchoring of spermatids to Sertoli cells caused a premature release of spermatids into the lumen. Our findings may help also explain the abnormal elongation process of remaining spermatids because these junctions and the correct positioning of germ cells in the epithelium are critically important for the progression of spermiogenesis. Interestingly, cell adhesion-related genes were generally upregulated in Dicer1 knockout germ cells. Claudin 5 ( Cldn5 ) was among the most upregulated genes and we show that the polarized localization of CLAUDIN5 in the apical ectoplasmic specializations was lost in Dicer1 knockout spermatids. Our results suggest that DICER-dependent pathways control the formation and organization of cell-cell junctions in the seminiferous epithelium via the regulation of cell adhesion-related genes.


Asunto(s)
Adhesión Celular/genética , ARN Helicasas DEAD-box/metabolismo , Uniones Intercelulares/metabolismo , Ribonucleasa III/metabolismo , Epitelio Seminífero/metabolismo , Células de Sertoli/metabolismo , Espermátides/metabolismo , Animales , Barrera Hematotesticular/metabolismo , Claudina-5/genética , Claudina-5/metabolismo , ARN Helicasas DEAD-box/genética , Uniones Intercelulares/genética , Masculino , Ratones , Ribonucleasa III/genética , Espermatogénesis/fisiología , Regulación hacia Arriba
11.
Reproduction ; 150(3): 209-16, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26245936

RESUMEN

Male fertility relies on the production of functional spermatozoa. Spermatogenesis is a complex differentiation process that is characterized by meiosis and dramatic morphogenesis of haploid cells. Spermatogenesis involves active changes in the microtubular network to support meiotic divisions, cell polarization, the reshaping of the nucleus, and the formation of a flagellum. Previously, we have demonstrated that a microtubule-based anterograde transport motor protein KIF3A is required for the sperm tail formation and nuclear shaping during spermatogenesis. In this study, we show that KIF3A interacts with a KIF1-binding protein (KBP) in the mouse testis. We have characterized the expression and localization pattern of KBP during spermatogenesis and localized both KIF3A and KBP in the cytoplasm of round spermatids and manchette of elongating spermatids. Interestingly, KBP localized also in the late chromatoid body (CB) of elongating spermatids, whose function involves intracellular movement and association with the microtubular network. Altogether our results suggest a role for KBP in spermatid elongation and in the function of the late CB.


Asunto(s)
Proteínas Portadoras/metabolismo , Haploidia , Cinesinas/metabolismo , Espermátides/metabolismo , Espermatogénesis , Testículo/metabolismo , Animales , Cinesinas/deficiencia , Cinesinas/genética , Masculino , Ratones Noqueados , Unión Proteica , Isoformas de Proteínas , Transducción de Señal , Testículo/citología
12.
Adv Exp Med Biol ; 888: 353-87, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26663192

RESUMEN

microRNAs constitute a large family of approximately 21-nucleotide-long, noncoding RNAs. They emerged more than 20 years ago as key posttranscriptional regulators of gene expression. The regulatory role of these small RNA molecules has recently begun to be explored in the human reproductive system. microRNAs have been shown to play an important role in control of reproductive functions, especially in the processes of oocyte maturation, folliculogenesis, corpus luteum function, implantation, and early embryonic development. Knockout of Dicer, the cytoplasmic enzyme that cleaves the pre-miRNA to its mature form, results in postimplantation embryonic lethality in several animal models, attributing to these small RNA vital functions in reproduction and development. Another intriguing characteristic of microRNAs is their presence in body fluids in a remarkably stable form that is protected from endogenous RNase activity. In this chapter we will describe the current knowledge on microRNAs, specifically relating to human gonadal cells. We will focus on their role in the ovarian physiologic process and ovulation dysfunction, regulation of spermatogenesis and male fertility, and putative involvement in human normal and aberrant trophoblast differentiation and invasion through the process of placentation.


Asunto(s)
MicroARNs/genética , Ovario/metabolismo , Ovulación/genética , Reproducción/genética , Espermatogénesis/genética , Animales , ARN Helicasas DEAD-box/genética , Femenino , Fertilidad/genética , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Masculino , Ovario/citología , Ovario/crecimiento & desarrollo , Síndrome del Ovario Poliquístico/genética , Ribonucleasa III/genética , Transducción de Señal/genética
13.
Subcell Biochem ; 61: 119-38, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23150249

RESUMEN

Male germ cell differentiation is a complex developmental program that produces highly specialized mature spermatozoa capable of independent movement and fertilization of an egg. Germ cells are unique in their capability to generate new organisms, and extra caution has to be taken to secure the correct inheritance of genetic and epigenetic information. Male germ cells are epigenetically distinct from somatic cells and they undergo several important epigenetic transitions. In primordial germ cells (PGCs), epigenome is reprogrammed by genome-wide resetting of epigenetic marks, including the sex-specific imprinting of certain genes. Postnatal spermatogenesis is characterized by drastic chromatin rearrangements during meiotic recombination, sex chromosome silencing, and compaction of sperm nuclei, which is accomplished by replacing near to all histones by sperm-specific protamines. Small RNAs, including microRNAs (miRNAs), endogenous small interfering RNAs (endo-siRNAs) and PIWI-interacting RNAs (piRNAs) are also involved in the control of male gamete production. The activities of small RNAs in male germ cells are diverse, and include miRNA- and endo-siRNA-mediated posttranscriptional mRNA regulation and piRNA-driven transposon silencing and the control of DNA methylation in PGCs. In this chapter, we give a brief review on the epigenetic processes that govern chromatin organization and germline-specific gene expression in differentiating male germ cells.


Asunto(s)
Epigénesis Genética , Espermatogénesis/genética , Espermatozoides/fisiología , Animales , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Regulación del Desarrollo de la Expresión Génica , Histonas/metabolismo , Humanos , Masculino , MicroARNs/metabolismo , Interferencia de ARN , Procesamiento Postranscripcional del ARN , ARN Interferente Pequeño
14.
Cell Rep ; 43(7): 114430, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38963760

RESUMEN

Cancer cells undergo major epigenetic alterations and transcriptomic changes, including ectopic expression of tissue- and cell-type-specific genes. Here, we show that the germline-specific RNA helicase DDX4 forms germ-granule-like cytoplasmic ribonucleoprotein granules in various human tumors, but not in cultured cancer cells. These cancerous DDX4 complexes contain RNA-binding proteins and splicing regulators, including many known germ granule components. The deletion of DDX4 in cancer cells induces transcriptomic changes and affects the alternative splicing landscape of a number of genes involved in cancer growth and invasiveness, leading to compromised capability of DDX4-null cancer cells to form xenograft tumors in immunocompromised mice. Importantly, the occurrence of DDX4 granules is associated with poor survival in patients with head and neck squamous cell carcinoma and higher histological grade of prostate cancer. Taken together, these results show that the germ-granule-resembling cancerous DDX4 granules control gene expression and promote malignant and invasive properties of cancer cells.

15.
Development ; 137(19): 3177-84, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20724452

RESUMEN

miR-18 belongs to the Oncomir-1 or miR-17~92 cluster that is intimately associated with the occurrence and progression of different types of cancer. However, the physiological roles of the Oncomir-1 cluster and its individual miRNAs are largely unknown. Here, we describe a novel function for miR-18 in mouse. We show that miR-18 directly targets heat shock factor 2 (HSF2), a transcription factor that influences a wide range of developmental processes including embryogenesis and gametogenesis. Furthermore, we show that miR-18 is highly abundant in testis, displaying distinct cell-type-specific expression during the epithelial cycle that constitutes spermatogenesis. Expression of HSF2 and of miR-18 exhibit an inverse correlation during spermatogenesis, indicating that, in germ cells, HSF2 is downregulated by miR-18. To investigate the in vivo function of miR-18 we developed a novel method, T-GIST, and demonstrate that inhibition of miR-18 in intact seminiferous tubules leads to increased HSF2 protein levels and altered expression of HSF2 target genes. Our results reveal that miR-18 regulates HSF2 activity in spermatogenesis and link miR-18 to HSF2-mediated physiological processes such as male germ cell maturation.


Asunto(s)
Proteínas de Choque Térmico/metabolismo , MicroARNs/genética , Espermatogénesis , Factores de Transcripción/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Línea Celular , Regulación de la Expresión Génica , Proteínas de Choque Térmico/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Espermatocitos/metabolismo , Factores de Transcripción/genética
16.
Semin Reprod Med ; 41(6): 213-225, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-38346711

RESUMEN

Male reproductive functions are strictly regulated in order to maintain sperm production and fertility. All processes are controlled by precise regulation of gene expression, which creates specific gene expression programs for different developmental stages and cell types, and forms the functional basis for the reproductive system. Small non-coding RNAs (sncRNAs) are involved in gene regulation by targeting mRNAs for translational repression and degradation through complementary base pairing to recognize their targets. This review article summarizes the current knowledge on the function of different classes of sncRNAs, in particular microRNAs (miRNAs) and PIWI-interacting RNAs (piRNAs), during male germ cell differentiation, with the focus on sncRNAs expressed in the germline. Although transcriptionally inactive, mature spermatozoa contain a complex population of sncRNAs, and we also discuss the recently identified role of sperm sncRNAs in the intergenerational transmission of epigenetic information on father's environmental and lifestyle exposures to offspring. Finally, we summarize the current information on the utility of sncRNAs as potential biomarkers of infertility that may aid in the diagnosis and prediction of outcomes of medically assisted reproduction.


Asunto(s)
MicroARNs , ARN Pequeño no Traducido , Humanos , Masculino , ARN Pequeño no Traducido/genética , ARN Pequeño no Traducido/metabolismo , Semen/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Espermatozoides/metabolismo , Reproducción/genética
17.
Andrology ; 11(4): 710-723, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36624638

RESUMEN

BACKGROUND: Germ granules are large cytoplasmic ribonucleoprotein complexes that emerge in the germline to participate in RNA regulation. The two most prominent germ granules are the intermitochondrial cement (IMC) in meiotic spermatocytes and the chromatoid body (CB) in haploid round spermatids, both functionally linked to the PIWI-interacting RNA (piRNA) pathway. AIMS: In this study, we clarified the IMC function by identifying proteins that form complexes with a well-known IMC protein PIWIL2/MILI in the mouse testis. RESULTS: The PIWIL2 interactome included several proteins with known functions in piRNA biogenesis. We further characterized the expression and localization of two of the identified proteins, Exonuclease 3'-5' domain-containing proteins EXD1 and EXD2, and confirmed their localization to the IMC. We showed that EXD2 interacts with PIWIL2, and that the mutation of Exd2 exonuclease domain in mice induces misregulation of piRNA levels originating from specific pachytene piRNA clusters, but does not disrupt male fertility. CONCLUSION: Altogether, this study highlights the central role of the IMC as a platform for piRNA biogenesis, and suggests that EXD1 and EXD2 function in the IMC-mediated RNA regulation in postnatal male germ cells.


Asunto(s)
ARN de Interacción con Piwi , Espermatocitos , Ratones , Masculino , Animales , Espermatogénesis/fisiología , Gránulos de Ribonucleoproteína de Células Germinales , Exonucleasas/metabolismo , Proteínas/metabolismo , ARN/metabolismo , ARN Interferente Pequeño/genética , Testículo/metabolismo
18.
Biomedicines ; 10(4)2022 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-35453658

RESUMEN

Excessive adiposity caused by high-fat diets (HFDs) is associated with testicular metabolic and functional abnormalities up to grand-offspring, but the mechanisms of this epigenetic inheritance are unclear. Here we describe an association of sperm small non-coding RNA (sncRNA) with testicular "inherited metabolic memory" of ancestral HFD, using a transgenerational rodent model. Male founders were fed a standard chow for 200 days (CTRL), HFD for 200 days (HFD), or standard chow for 60 days followed by HFD for 140 days (HFDt). The male offspring and grand-offspring were fed standard chow for 200 days. The sncRNA sequencing from epidydimal spermatozoa revealed signatures associated with testicular metabolic plasticity in HFD-exposed mice and in the unexposed progeny. Sperm tRNA-derived RNA (tsRNA) and repeat-derived small RNA (repRNA) content were specially affected by HFDt and in the offspring of HFD and HFDt mice. The grand-offspring of HFD and HFDt mice showed lower sperm counts than CTRL descendants, whereas the sperm miRNA content was affected. Although the causality between sperm sncRNAs content and transgenerational epigenetic inheritance of HFD-related traits remains elusive, our results suggest that sperm sncRNA content is influenced by ancestral exposure to HFD, contributing to the sperm epigenome up to the grand-offspring.

19.
Endocrinology ; 164(2)2022 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-36461763

RESUMEN

Spermatogenesis is a complex differentiation process that takes place in the seminiferous tubules. A specific organization of spermatogenic cells within the seminiferous epithelium enables a synchronous progress of germ cells at certain steps of differentiation on the spermatogenic pathway. This can be observed in testis cross-sections where seminiferous tubules can be classified into distinct stages of constant cellular composition (12 stages in the mouse). For a detailed analysis of spermatogenesis, these stages have to be individually observed from testis cross-sections. However, the recognition of stages requires special training and expertise. Furthermore, the manual scoring is laborious considering the high number of tubule cross-sections that have to be analyzed. To facilitate the analysis of spermatogenesis, we have developed a convolutional deep neural network-based approach named "STAGETOOL." STAGETOOL analyses histological images of 4',6-diamidine-2'-phenylindole dihydrochloride (DAPI)-stained mouse testis cross-sections at ×400 magnification, and very accurately classifies tubule cross-sections into 5 stage classes and cells into 9 categories. STAGETOOL classification accuracy for stage classes of seminiferous tubules of a whole-testis cross-section is 99.1%. For cellular level analysis the F1 score for 9 seminiferous epithelial cell types ranges from 0.80 to 0.98. Furthermore, we show that STAGETOOL can be applied for the analysis of knockout mouse models with spermatogenic defects, as well as for automated profiling of protein expression patterns. STAGETOOL is the first fluorescent labeling-based automated method for mouse testis histological analysis that enables both stage and cell-type recognition. While STAGETOOL qualitatively parallels an experienced human histologist, it outperforms humans time-wise, therefore representing a major advancement in male reproductive biology research.


Asunto(s)
Túbulos Seminíferos , Testículo , Masculino , Ratones , Humanos , Animales , Espermatogénesis , Epitelio Seminífero , Células Epiteliales
20.
Biol Reprod ; 85(4): 690-701, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21715716

RESUMEN

Primary ciliary dyskinesia (PCD) results from defects in motile cilia function. Mice homozygous for the mutation big giant head (bgh) have several abnormalities commonly associated with PCD, including hydrocephalus, male infertility, and sinusitis. In the present study, we use a variety of histopathological and cell biological techniques to characterize the bgh phenotype, and we identify the bgh mutation using a positional cloning approach. Histopathological, immunofluorescence, and electron microscopic analyses demonstrate that the male infertility results from shortened flagella and disorganized axonemal and accessory structures in elongating spermatids and mature sperm. In addition, there is a reduced number of elongating spermatids during spermatogenesis and mature sperm in the epididymis. Histological analyses show that the hydrocephalus is characterized by severe dilatation of the lateral ventricles and that bgh sinuses have an accumulation of mucus infiltrated by neutrophils. In contrast to the sperm phenotype, electron microscopy demonstrates that mutant respiratory epithelial cilia are ultrastructurally normal, but video microscopic analysis shows that their beat frequency is lower than that of wild-type cilia. Through a positional cloning approach, we identified two sequence variants in the gene encoding sperm flagellar protein 2 (SPEF2), which has been postulated to play an important role in spermatogenesis and flagellar assembly. A causative nonsense mutation was validated by Western blot analysis, strongly suggesting that the bgh phenotype results from the loss of SPEF2 function. Taken together, the data in this study demonstrate that SPEF2 is required for cilia function and identify a new genetic cause of PCD in mice.


Asunto(s)
Trastornos de la Motilidad Ciliar/fisiopatología , Infertilidad Masculina/patología , Proteínas/fisiología , Animales , Secuencia de Bases , Trastornos de la Motilidad Ciliar/patología , Epidídimo/metabolismo , Epidídimo/ultraestructura , Hidrocefalia/fisiopatología , Infertilidad Masculina/metabolismo , Masculino , Sinusitis Maxilar/inmunología , Sinusitis Maxilar/patología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Datos de Secuencia Molecular , Infiltración Neutrófila , Proteínas/química , Proteínas/genética , Mucosa Respiratoria/fisiopatología , Mucosa Respiratoria/ultraestructura , Cola del Espermatozoide/metabolismo , Cola del Espermatozoide/ultraestructura , Espermátides/metabolismo , Espermátides/ultraestructura , Espermatogénesis , Espermatozoides/metabolismo , Espermatozoides/ultraestructura , Tráquea/fisiopatología , Tráquea/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA