Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 337
Filtrar
1.
Development ; 151(8)2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38691389

RESUMEN

Mammalian spermatogenesis, probably the most complex of all cellular developmental processes, is an ideal model both for studying the specific mechanism of gametogenesis and for understanding the basic rules governing all developmental processes, as it entails both cell type-specific and housekeeping molecular processes. Spermatogenesis can be viewed as a mission with many tasks to accomplish, and its success is genetically programmed and ensured by the collaboration of a large number of genes. Here, I present an overview of mammalian spermatogenesis and the mechanisms underlying each step in the process, covering the cellular and molecular activities that occur at each developmental stage and emphasizing their gene regulation in light of recent studies.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Espermatogénesis , Animales , Humanos , Masculino , Mamíferos/genética
2.
Development ; 150(9)2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-37082969

RESUMEN

Unique chromatin remodeling factors orchestrate dramatic changes in nuclear morphology during differentiation of the mature sperm head. A crucial step in this process is histone-to-protamine exchange, which must be executed correctly to avoid sperm DNA damage, embryonic lethality and male sterility. Here, we define an essential role for the histone methyltransferase DOT1L in the histone-to-protamine transition. We show that DOT1L is abundantly expressed in mouse meiotic and postmeiotic germ cells, and that methylation of histone H3 lysine 79 (H3K79), the modification catalyzed by DOT1L, is enriched in developing spermatids in the initial stages of histone replacement. Elongating spermatids lacking DOT1L fail to fully replace histones and exhibit aberrant protamine recruitment, resulting in deformed sperm heads and male sterility. Loss of DOT1L results in transcriptional dysregulation coinciding with the onset of histone replacement and affecting genes required for histone-to-protamine exchange. DOT1L also deposits H3K79me2 and promotes accumulation of elongating RNA Polymerase II at the testis-specific bromodomain gene Brdt. Together, our results indicate that DOT1L is an important mediator of transcription during spermatid differentiation and an indispensable regulator of male fertility.


Asunto(s)
Histonas , Espermátides , Animales , Masculino , Ratones , Diferenciación Celular/genética , Ensamble y Desensamble de Cromatina , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Protaminas/genética , Protaminas/metabolismo , Semen/metabolismo , Espermátides/metabolismo
3.
Development ; 150(13)2023 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-37283046

RESUMEN

In mammals, a near complete resetting of DNA methylation (DNAme) is observed during germline establishment. This wave of epigenetic reprogramming is sensitive to the environment, which could impair the establishment of an optimal state of the gamete epigenome, hence proper embryo development. Yet, we lack a comprehensive understanding of DNAme dynamics during spermatogenesis, especially in rats, the model of choice for toxicological studies. Using a combination of cell sorting and DNA methyl-seq capture, we generated a stage-specific mapping of DNAme in nine populations of differentiating germ cells from perinatal life to spermiogenesis. DNAme was found to reach its lowest level at gestational day 18, the last demethylated coding regions being associated with negative regulation of cell movement. The following de novo DNAme displayed three different kinetics with common and distinct genomic enrichments, suggesting a non-random process. DNAme variations were also detected at key steps of chromatin remodeling during spermiogenesis, revealing potential sensitivity. These methylome datasets for coding sequences during normal spermatogenesis in rat provide an essential reference for studying epigenetic-related effects of disease or environmental factors on the male germline.


Asunto(s)
Metilación de ADN , Células Germinativas , Masculino , Embarazo , Femenino , Ratas , Animales , Metilación de ADN/genética , Espermatogénesis/genética , ADN , Epigenoma , Mamíferos/genética
4.
Development ; 150(9)2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-37082953

RESUMEN

Histone modifications regulate chromatin remodeling and gene expression in development and diseases. DOT1L, the sole histone H3K79 methyltransferase, is essential for embryonic development. Here, we report that DOT1L regulates male fertility in mouse. DOT1L associates with MLLT10 in testis. DOT1L and MLLT10 localize to the sex chromatin in meiotic and post-meiotic germ cells in an inter-dependent manner. Loss of either DOT1L or MLLT10 leads to reduced testis weight, decreased sperm count and male subfertility. H3K79me2 is abundant in elongating spermatids, which undergo the dramatic histone-to-protamine transition. Both DOT1L and MLLT10 are essential for H3K79me2 modification in germ cells. Strikingly, histones are substantially retained in epididymal sperm from either DOT1L- or MLLT10-deficient mice. These results demonstrate that H3K79 methylation promotes histone replacement during spermiogenesis.


Asunto(s)
Histonas , Semen , Animales , Masculino , Ratones , Fertilidad , Histona Metiltransferasas/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Metilación , Metiltransferasas/genética , Semen/metabolismo , Espermatogénesis/genética , Factores de Transcripción/metabolismo
5.
Development ; 150(21)2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37882665

RESUMEN

Sperm flagellum plays a crucial role in male fertility. Here, we generated Ccdc183 knockout mice using the CRISPR/Cas9 system to reveal the protein function of the testis-specific protein CCDC183 in spermiogenesis. We demonstrated that the absence of CCDC183 causes male infertility with morphological and motility defects in spermatozoa. Owing to the lack of CCDC183, centrioles after elongation of axonemal microtubules do not connect the cell surface and nucleus during spermiogenesis, which causes subsequent loss of cytoplasmic invagination around the flagellum. As a result, the flagellar compartment does not form properly and cytosol-exposed axonemal microtubules collapse during spermiogenesis. In addition, ectopic localization of accessory structures, such as the fibrous sheath and outer dense fibers, and abnormal head shape as a result of abnormal sculpting by the manchette are observed in Ccdc183 knockout spermatids. Our results indicate that CCDC183 plays an essential role in cytoplasmic invagination around the flagellum to form functional spermatozoa during spermiogenesis.


Asunto(s)
Semen , Espermatogénesis , Ratones , Animales , Masculino , Citosol , Espermatogénesis/genética , Flagelos , Ratones Noqueados , Fertilidad/genética
6.
Proc Natl Acad Sci U S A ; 120(45): e2313787120, 2023 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-37903275

RESUMEN

The manchette is a crucial transient structure involved in sperm development, with its composition and regulation still not fully understood. This study focused on investigating the roles of CAMSAP1 and CAMSAP2, microtubule (MT) minus-end binding proteins, in regulating manchette MTs, spermiogenesis, and male fertility. The loss of CAMSAP1, but not CAMSAP2, disrupts the well-orchestrated process of spermiogenesis, leading to abnormal manchette elongation and delayed removal, resulting in deformed sperm nuclei and tails resembling oligoasthenozoospermia symptoms. We investigated the underlying molecular mechanisms by purifying manchette assemblies and comparing them through proteomic analysis, and results showed that the absence of CAMSAP1 disrupted the proper localization of key proteins (CEP170 and KIF2A) at the manchette minus end, compromising its structural integrity and hindering MT depolymerization. These findings highlight the significance of maintaining homeostasis in manchette MT minus-ends for shaping manchette morphology during late spermiogenesis, offering insights into the molecular mechanisms underlying infertility and sperm abnormalities.


Asunto(s)
Proteómica , Semen , Humanos , Masculino , Espermatogénesis/fisiología , Microtúbulos/metabolismo , Fertilidad
7.
J Cell Sci ; 136(7)2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36825599

RESUMEN

SUN domain proteins are conserved proteins of the nuclear envelope and key components of the LINC complexes (for 'linkers of the nucleoskeleton and the cytoskeleton'). Previous studies have demonstrated that the testis-specific SUN domain protein SUN4 (also known as SPAG4) is a vital player in the directed shaping of the spermatid nucleus. However, its molecular properties relating to this crucial function have remained largely unknown, and controversial data for the organization and orientation of SUN4 within the spermatid nuclear envelope have been presented so far. Here, we have re-evaluated this issue in detail and show robust evidence that SUN4 is integral to the inner nuclear membrane, sharing a classical SUN domain protein topology. The C-terminal SUN domain of SUN4 localizes to the perinuclear space, whereas the N-terminus is directed to the nucleoplasm, interacting with the spermiogenesis-specific lamin B3. We found that SUN4 forms heteromeric assemblies with SUN3 in vivo and regulates SUN3 expression. Together, our results contribute to a better understanding of the specific function of SUN4 at the spermatid nucleo-cytoplasmic junction and the process of sperm-head formation.


Asunto(s)
Membrana Nuclear , Espermátides , Humanos , Masculino , Proteínas de la Membrana/metabolismo , Membrana Nuclear/metabolismo , Semen/metabolismo , Espermátides/metabolismo , Proteínas Nucleares/metabolismo , Lamina Tipo B
8.
Development ; 149(15)2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35924955

RESUMEN

Gametogenesis is an essential event for sexual reproduction in various organisms. Bryophytes employ motile sperm (spermatozoids) as male gametes, which locomote to the egg cells to accomplish fertilization. The spermatozoids of bryophytes harbor distinctive morphological characteristics, including a cell body with a helical shape and two flagella. During spermiogenesis, the shape and cellular contents of the spermatids are dynamically reorganized. However, the reorganization patterns of each organelle remain obscure. In this study, we classified the developmental processes during spermiogenesis in the liverwort Marchantia polymorpha according to changes in cellular and nuclear shapes and flagellar development. We then examined the remodeling of microtubules and the reorganization of endomembrane organelles. The results indicated that the state of glutamylation of tubulin changes during formation of the flagella and spline. We also found that the plasma membrane and endomembrane organelles are drastically reorganized in a precisely regulated manner, which involves the functions of endosomal sorting complexes required for transport (ESCRT) machineries in endocytic and vacuolar transport. These findings are expected to provide useful indices to classify developmental and subcellular processes of spermiogenesis in bryophytes.


Asunto(s)
Marchantia , Núcleo Celular , Marchantia/metabolismo , Microtúbulos/metabolismo , Semillas , Espermatogénesis
9.
Development ; 149(12)2022 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-35588208

RESUMEN

As one of the post-transcriptional regulatory mechanisms, uncoupling of transcription and translation plays an essential role in development and adulthood physiology. However, it remains elusive how thousands of mRNAs get translationally silenced while stability is maintained for hours or even days before translation. In addition to oocytes and neurons, developing spermatids display significant uncoupling of transcription and translation for delayed translation. Therefore, spermiogenesis represents an excellent in vivo model for investigating the mechanism underlying uncoupled transcription and translation. Through full-length poly(A) deep sequencing, we discovered dynamic changes in poly(A) length through deadenylation and re-polyadenylation. Deadenylation appeared to be mediated by microRNAs (miRNAs), and transcripts with shorter poly(A) tails tend to be sequestered into ribonucleoprotein (RNP) granules for translational repression and stabilization. In contrast, re-polyadenylation might allow for translocation of the translationally repressed transcripts from RNP granules to polysomes. Overall, our data suggest that miRNA-dependent poly(A) length control represents a previously unreported mechanism underlying uncoupled translation and transcription in haploid male mouse germ cells.


Asunto(s)
MicroARNs , Poli A , Animales , Haploidia , Masculino , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Poli A/metabolismo , Biosíntesis de Proteínas , ARN Mensajero/metabolismo , Espermátides/metabolismo
10.
Development ; 149(18)2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-35993297

RESUMEN

Round spermatid injection (ROSI) results in a lower birth rate than intracytoplasmic sperm injection, which has hampered its clinical application. Inefficient development of ROSI embryos has been attributed to epigenetic abnormalities. However, the chromatin-based mechanism that underpins the low birth rate in ROSI remains to be determined. Here, we show that a repressive histone mark, H3K27me3, persists from mouse round spermatids into zygotes in ROSI and that round spermatid-derived H3K27me3 is associated with less accessible chromatin and impaired gene expression in ROSI embryos. These loci are initially marked by H3K27me3 but undergo histone modification remodelling in spermiogenesis, resulting in reduced H3K27me3 in normal spermatozoa. Therefore, the absence of epigenetic remodelling, presumably mediated by histone turnover during spermiogenesis, leads to dysregulation of chromatin accessibility and transcription in ROSI embryos. Thus, our results unveil a molecular logic, in which chromatin states in round spermatids impinge on chromatin accessibility and transcription in ROSI embryos, highlighting the importance of epigenetic remodelling during spermiogenesis in successful reproduction.


Asunto(s)
Histonas , Espermátides , Animales , Cromatina/genética , Cromatina/metabolismo , Histonas/genética , Histonas/metabolismo , Masculino , Ratones , Oocitos/metabolismo , Herencia Paterna , Semen/metabolismo , Espermátides/metabolismo
11.
Development ; 149(16)2022 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-35950913

RESUMEN

Profilin 4 (Pfn4) is expressed during spermiogenesis and localizes to the acrosome-acroplaxome-manchette complex. Here, we generated PFN4-deficient mice, with sperm displaying severe impairment in manchette formation. Interestingly, HOOK1 staining suggests that the perinuclear ring is established; however, ARL3 staining is disrupted, suggesting that lack of PFN4 does not interfere with the formation of the perinuclear ring and initial localization of HOOK1, but impedes microtubular organization of the manchette. Furthermore, amorphous head shape and flagellar defects were detected, resulting in reduced sperm motility. Disrupted cis- and trans-Golgi networks and aberrant production of proacrosomal vesicles caused impaired acrosome biogenesis. Proteomic analysis showed that the proteins ARF3, SPECC1L and FKBP1, which are involved in Golgi membrane trafficking and PI3K/AKT pathway, are more abundant in Pfn4-/- testes. Levels of PI3K, AKT and mTOR were elevated, whereas AMPK level was reduced, consistent with inhibition of autophagy. This seems to result in blockage of autophagic flux, which could explain the failure in acrosome formation. In vitro fertilization demonstrated that PFN4-deficient sperm is capable of fertilizing zona-free oocytes, suggesting a potential treatment for PFN4-related human infertility.


Asunto(s)
Acrosoma , Profilinas , Espermátides , Espermatogénesis , Acrosoma/metabolismo , Animales , Masculino , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Profilinas/genética , Profilinas/metabolismo , Proteómica , Proteínas Proto-Oncogénicas c-akt/metabolismo , Semen , Motilidad Espermática , Espermátides/metabolismo , Espermatogénesis/genética , Espermatozoides
12.
Mol Cell ; 66(1): 89-101.e8, 2017 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-28366643

RESUMEN

Histone replacement by transition proteins (TPs) and protamines (Prms) constitutes an essential step for the successful production of functional male gametes, yet nothing is known on the underlying functional interplay between histones, TPs, and Prms. Here, by studying spermatogenesis in the absence of a spermatid-specific histone variant, H2A.L.2, we discover a fundamental mechanism involved in the transformation of nucleosomes into nucleoprotamines. H2A.L.2 is synthesized at the same time as TPs and enables their loading onto the nucleosomes. TPs do not displace histones but rather drive the recruitment and processing of Prms, which are themselves responsible for histone eviction. Altogether, the incorporation of H2A.L.2 initiates and orchestrates a series of successive transitional states that ultimately shift to the fully compacted genome of the mature spermatozoa. Hence, the current view of histone-to-nucleoprotamine transition should be revisited and include an additional step with H2A.L.2 assembly prior to the action of TPs and Prms.


Asunto(s)
Ensamble y Desensamble de Cromatina , Cromatina/metabolismo , Histonas/metabolismo , Nucleosomas/metabolismo , Protaminas/metabolismo , Espermatogénesis , Espermatozoides/metabolismo , Animales , Células COS , Chlorocebus aethiops , Cromatina/genética , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Biología Computacional , Bases de Datos Genéticas , Fertilidad , Regulación del Desarrollo de la Expresión Génica , Predisposición Genética a la Enfermedad , Genoma , Histonas/deficiencia , Histonas/genética , Infertilidad Masculina/genética , Infertilidad Masculina/metabolismo , Infertilidad Masculina/patología , Infertilidad Masculina/fisiopatología , Masculino , Ratones de la Cepa 129 , Ratones Noqueados , Nucleosomas/genética , Fenotipo , Espermatogénesis/genética , Espermatozoides/patología , Transfección
13.
Semin Cell Dev Biol ; 121: 125-132, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34325997

RESUMEN

Studies have demonstrated that biologically active fragments are generated from the basement membrane and the Sertoli cell-spermatid adhesion site known as apical ectoplasmic specialization (apical ES, a testis-specific actin-based anchoring junction) in the rat testis. These bioactive fragments or peptides are produced locally across the seminiferous epithelium through proteolytic cleavage of constituent proteins at the basement membrane and the apical ES. Studies have shown that they are being used to modulate and coordinate cellular functions across the seminiferous epithelium during different stages of the epithelial cycle of spermatogenesis. In this review, we briefly summarize recent findings based on studies using rat testes as a study model regarding the role of these bioactive peptides that serve as a local regulatory network to support spermatogenesis. We also used scRNA-Seq transcriptome datasets in the public domain for OA (obstructive azoospermia) and NAO (non-obstructive azoospermia) human testes versus testes from normal men for analysis in this review. It was shown that there are differential expression of different collagen chains and laminin chains in these testes, suggesting the possibility of a similar local regulatory network in the human testis to support spermatogenesis, and the possible disruption of such network in men is associated with OA and/or NOA.


Asunto(s)
Colágeno/metabolismo , Perfilación de la Expresión Génica/métodos , Laminina/metabolismo , Análisis de la Célula Individual/métodos , Espermatogénesis/genética , Animales , Humanos , Masculino , Ratones , Persona de Mediana Edad , Ratas
14.
Dev Biol ; 504: 137-148, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37805103

RESUMEN

Spermiogenesis, a sperm-activation step, is crucial for the transformation of immotile spermatids into motile sperm. Though membrane transport of ions and molecules across the sperm plasma membrane has been implicated in this process, the full repertoire of transporters involved, and their respective substrates, is unclear. Here, we report that the major facilitator superfamily transporter SPIN-4/Spinster governs efficient spermiogenesis and fertility in the hermaphrodite nematode Caenorhabditis elegans. Unlike other C. elegans Spinster paralogs, SPIN-4 is germline-expressed. Moreover, SPIN-4 expression is gamete-specific; it is strongly expressed in developing sperm, where it localizes to the plasma membrane, but it is absent from oocytes. Consistent with these expression data, we demonstrate that knocking out spin-4 impairs sperm development, leading to the formation of non-motile sperm that lack pseudopodia. Consequently, hermaphrodites homozygous for the spin-4(knu1099) knockout allele show extensive sperm wasting and reduced self-progeny. We observe similar defects when we genetically inhibit production of sphingosine-1-phosphate, a lipid molecule that stimulates cell motility when exported extracellularly by Spinster homologs in other contexts. Remarkably, extracellular supplementation with sphingosine-1-phosphate rescues sperm activation and motility in the absence of SPIN-4, suggesting that Spinster-dependent efflux of sphingosine-1-phosphate plays a key role in sperm mobilization. These findings identify a new signaling mechanism in C. elegans spermiogenesis entailing Spinster and sphingosine-1-phosphate.


Asunto(s)
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Animales , Masculino , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Semen/metabolismo , Espermatozoides/metabolismo
15.
J Cell Sci ; 135(3)2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34931239

RESUMEN

Transcription factor-like 5 (TCFL5) is a testis-specific protein that contains the basic helix-loop-helix domain, but the in vivo functions of TCFL5 remain unknown. Herein, we generated CRISPR/Cas9-mediated knockout mice to dissect the function of TCFL5 in mouse testes. Surprisingly, we found that it was difficult to generate homozygous mice with the Tcfl5 deletion as the heterozygous males (Tcfl5+/-) were infertile. However, we did observe markedly abnormal phenotypes of spermatids and spermatozoa in the testes and epididymides of Tcfl5+/- mice. Mechanistically, we demonstrated that TCFL5 transcriptionally and post-transcriptionally regulated a set of genes participating in male germ cell development via TCFL5 ChIP-DNA and eCLIP-RNA high-throughput sequencing. We also identified a known RNA-binding protein, FXR1, as an interacting partner of TCFL5 that may coordinate the transition and localization of TCFL5 in the nucleus. Collectively, we herein report for the first time that Tcfl5 is haploinsufficient in vivo and acts as a dual-function protein that mediates DNA and RNA to regulate spermatogenesis. This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Espermatogénesis , Testículo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , ADN/metabolismo , Fertilidad/genética , Masculino , Ratones , Ratones Noqueados , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Espermatogénesis/genética , Espermatozoides/metabolismo , Testículo/metabolismo , Factores de Transcripción/metabolismo
16.
Development ; 148(11)2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34100066

RESUMEN

Spermatogenesis is precisely controlled by complex gene-expression programs. During mammalian male germ-cell development, a crucial feature is the repression of transcription before spermatid elongation. Previously, we discovered that the RNA-binding protein EWSR1 plays an important role in meiotic recombination in mouse, and showed that EWSR1 is highly expressed in late meiotic cells and post-meiotic cells. Here, we used an Ewsr1 pachytene stage-specific knockout mouse model to study the roles of Ewsr1 in late meiotic prophase I and in spermatozoa maturation. We show that loss of EWSR1 in late meiotic prophase I does not affect proper meiosis completion, but does result in defective spermatid elongation and chromocenter formation in the developing germ cells. As a result, male mice lacking EWSR1 after pachynema are sterile. We found that, in Ewsr1 CKO round spermatids, transition from a meiotic gene-expression program to a post-meiotic and spermatid gene expression program related to DNA condensation is impaired, suggesting that EWSR1 plays an important role in regulation of spermiogenesis-related mRNA synthesis necessary for spermatid differentiation into mature sperm.


Asunto(s)
Proteína EWS de Unión a ARN/genética , Proteína EWS de Unión a ARN/metabolismo , Espermátides/metabolismo , Espermatogénesis/genética , Espermatogénesis/fisiología , Animales , Regulación del Desarrollo de la Expresión Génica , Masculino , Meiosis , Profase Meiótica I , Ratones , Ratones Noqueados , Espermatozoides
17.
Development ; 148(24)2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34822718

RESUMEN

Katanin microtubule-severing enzymes are crucial executers of microtubule regulation. Here, we have created an allelic loss-of-function series of the katanin regulatory B-subunit KATNB1 in mice. We reveal that KATNB1 is the master regulator of all katanin enzymatic A-subunits during mammalian spermatogenesis, wherein it is required to maintain katanin A-subunit abundance. Our data shows that complete loss of KATNB1 from germ cells is incompatible with sperm production, and we reveal multiple new spermatogenesis functions for KATNB1, including essential roles in male meiosis, acrosome formation, sperm tail assembly, regulation of both the Sertoli and germ cell cytoskeletons during sperm nuclear remodelling, and maintenance of seminiferous epithelium integrity. Collectively, our findings reveal that katanins are able to differentially regulate almost all key microtubule-based structures during mammalian male germ cell development, through the complexing of one master controller, KATNB1, with a 'toolbox' of neofunctionalised katanin A-subunits.


Asunto(s)
Haploidia , Katanina/genética , Meiosis/genética , Espermatogénesis/genética , Espermatozoides/crecimiento & desarrollo , Acrosoma/metabolismo , Animales , Citoesqueleto/genética , Células Germinativas/citología , Células Germinativas/crecimiento & desarrollo , Masculino , Ratones , Microtúbulos/genética , Células de Sertoli/citología , Cola del Espermatozoide/metabolismo , Espermatozoides/metabolismo
18.
Biol Reprod ; 110(3): 599-614, 2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-37975917

RESUMEN

Mammalian spermatogenesis is a highly complex multi-step biological process, and autophagy has been demonstrated to be involved in the process of spermatogenesis. Beclin-1/BECN1, a core autophagy factor, plays a critical role in many biological processes and diseases. However, its function in spermatogenesis remains largely unclear. In the present study, germ cell-specific Beclin 1 (Becn1) knockout mice were generated and were conducted to determine the role of Becn1 in spermatogenesis and fertility of mice. Results indicate that Becn1 deficiency leads to reduced sperm motility and quantity, partial failure of spermiation, actin network disruption, excessive residual cytoplasm, acrosome malformation, and aberrant mitochondrial accumulation of sperm, ultimately resulting in reduced fertility in male mice. Furthermore, inhibition of autophagy was observed in the testes of germ cell-specific Becn1 knockout mice, which may contribute to impaired spermiogenesis and reduced fertility. Collectively, our results reveal that Becn1 is essential for fertility and spermiogenesis in mice.


Asunto(s)
Infertilidad Masculina , Animales , Humanos , Masculino , Ratones , Autofagia , Beclina-1/genética , Beclina-1/metabolismo , Fertilidad/genética , Infertilidad Masculina/metabolismo , Mamíferos , Ratones Noqueados , Semen/metabolismo , Motilidad Espermática/genética , Espermatogénesis/genética , Espermatozoides/metabolismo
19.
Biol Reprod ; 2024 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-39018224

RESUMEN

In male reproductive system, proteins containing the coiled-coil domain (CCDC) are predominantly expressed in specific regions including the testis, epididymis, seminal vesicle, and prostate. They play a vital role in centriole formation, sperm motility and flagellar development in male gametes. Despite being highly expressed in the testis, the exact physiological function of the coiled-coil domain-containing 189 (Ccdc189) gene remain largely unclear. Our research provides a comprehensive and detailed investigation into the localization of CCDC189 protein within the testis seminiferous tubules. CCDC189 specifically expressed in spermatocytes, round spermatids and elongating spermatids in mouse testis. The deletion of Ccdc189 in mouse leads to male infertility, characterized by significantly reduced sperm counts and motility. Abnormally shaped spermatozoa with irregular tails, exhibiting shortened and twisted morphology, were observed in the seminiferous tubules. Electron microscopy revealed disordered and missing peripheral microtubule doublets (MTD) and outer dense fibers (ODF) in the sperm flagella, accompanied by a consistent absence of central pairs (CP). The knockout of Ccdc189 resulted in oligo-astheno-teratozoospermia, which is characterized by low sperm count and reduced sperm motility and abnormal morphology. Furthermore, we identified poly(A)-binding protein cytoplasmic 1 (PABPC1) and PABPC2 as interacting proteins with CCDC189. These proteins belong to the poly(A)-binding protein (PABP) family and are involved in regulating mRNA translational activity in spermatogenic cells by specifically binding to poly(A) tails at the 3' ends of mRNAs.

20.
Cell Mol Life Sci ; 81(1): 1, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-38038747

RESUMEN

Multiple morphological abnormalities of the flagella (MMAF) is a severe disease of male infertility, while the pathogenetic mechanisms of MMAF are still incompletely understood. Previously, we found that the deficiency of Ccdc38 might be associated with MMAF. To understand the underlying mechanism of this disease, we identified the potential partner of this protein and found that the coiled-coil domain containing 146 (CCDC146) can interact with CCDC38. It is predominantly expressed in the testes, and the knockout of this gene resulted in complete infertility in male mice but not in females. The knockout of Ccdc146 impaired spermiogenesis, mainly due to flagellum and manchette organization defects, finally led to MMAF-like phenotype. Furthermore, we demonstrated that CCDC146 could interact with both CCDC38 and CCDC42. It also interacts with intraflagellar transport (IFT) complexes IFT88 and IFT20. The knockout of this gene led to the decrease of ODF2, IFT88, and IFT20 protein levels, but did not affect CCDC38, CCDC42, or ODF1 expression. Additionally, we predicted and validated the detailed interactions between CCDC146 and CCDC38 or CCDC42, and built the interaction models at the atomic level. Our results suggest that the testis predominantly expressed gene Ccdc146 is essential for sperm flagellum biogenesis and male fertility, and its mutations might be associated with MMAF in some patients.


Asunto(s)
Infertilidad Masculina , Proteínas Asociadas a Microtúbulos , Cola del Espermatozoide , Animales , Masculino , Ratones , Fertilidad/genética , Proteínas de Choque Térmico/metabolismo , Infertilidad Masculina/metabolismo , Ratones Noqueados , Semen , Cola del Espermatozoide/metabolismo , Cola del Espermatozoide/patología , Espermatozoides/metabolismo , Testículo/metabolismo , Proteínas Asociadas a Microtúbulos/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda