Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 518
Filter
1.
Fly (Austin) ; 17(1): 2234795, 2023 12.
Article in English | MEDLINE | ID: mdl-37436409

ABSTRACT

Valosin-containing protein (VCP) is a versatile and ubiquitously expressed AAA+ ATPase that regulates multiple stages of Drosophila spermatogenesis. While VCP has documented roles in mitotic spermatogonia and meiotic spermatocytes, it is also highly expressed in post-meiotic spermatids, suggesting potential late-stage developmental functions as well. However, tools to assess late-stage activities of pleiotropic spermatogenesis genes such as VCP are lacking. Available germline-specific Gal4 drivers activate in stem cells or spermatogonia; consequently, knocking down VCP using one of these drivers disrupts or blocks early germ-cell development, precluding analysis of VCP in later stages. A Gal4 driver that activates later in development, such as at the meiotic spermatocyte stage, may permit functional analyses of VCP and other factors in post-meiotic stages. Here, we describe a germline-specific Gal4 driver, Rbp4-Gal4, which drives transgene expression beginning in the early spermatocyte stage. We find that Rbp4-Gal4-driven knockdown of VCP causes defects in spermatid chromatin condensation and individualization without affecting earlier developmental stages. Interestingly, the defect in chromatin condensation appears linked to errors in the histone-to-protamine transition, a key event in spermatid development. Overall, our study reveals roles for VCP in spermatid development and establishes a powerful tool to dissect the functions of pleiotropic spermatogenesis genes.


Subject(s)
Spermatids , Spermatogenesis , Male , Animals , Spermatids/physiology , Valosin Containing Protein/genetics , Spermatogenesis/genetics , Meiosis , Drosophila/genetics , Chromatin
2.
Reprod Domest Anim ; 57(5): 459-464, 2022 May.
Article in English | MEDLINE | ID: mdl-35098603

ABSTRACT

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


Subject(s)
Spermatids , Spermatogenesis , Animals , Cats , Male , Mammals , Meiosis , Spermatids/physiology , Spermatocytes/physiology , Spermatogenesis/physiology , Spermatogonia , Spermatozoa/physiology , Testis
3.
Reprod Biol Endocrinol ; 19(1): 144, 2021 Sep 15.
Article in English | MEDLINE | ID: mdl-34526015

ABSTRACT

In mammals, germline development undergoes dramatic morphological and molecular changes and is epigenetically subject to intricate yet exquisite regulation. Which epigenetic players and how they participate in the germline developmental process are not fully characterized. Spin1 is a multifunctional epigenetic protein reader that has been shown to recognize H3 "K4me3-R8me2a" histone marks, and more recently the non-canonical bivalent H3 "K4me3-K9me3/2" marks as well. As a robust Spin1-interacting cofactor, Spindoc has been identified to enhance the binding of Spin1 to its substrate histone marks, thereby modulating the downstream signaling; However, the physiological role of Spindoc in germline development is unknown. We generated two Spindoc knockout mouse models through CRISPR/Cas9 strategy, which revealed that Spindoc is specifically required for haploid spermatid development, but not essential for meiotic divisions in spermatocytes. This study unveiled a new epigenetic player that participates in haploid germline development.


Subject(s)
Co-Repressor Proteins , Spermatids/physiology , Spermatogenesis/genetics , Animals , Cell Cycle Proteins/metabolism , Cell Differentiation/genetics , Cell Division/genetics , Co-Repressor Proteins/genetics , Co-Repressor Proteins/metabolism , Haploidy , Male , Meiosis/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Phosphoproteins/metabolism , Protein Binding
4.
Gene ; 798: 145795, 2021 Sep 25.
Article in English | MEDLINE | ID: mdl-34175396

ABSTRACT

Spermiogenesis is the final phase of spermatogenesis, wherein the spermatids differentiate into mature spermatozoa via complex morphological transformation. In this process, kinesin plays an important role. Here, we observed the morphological transformation of spermatids and analyzed the characterization, dynamic transcription, and potential function of kinesin KIF3A/KIF3B during spermiogenesis in Chinese hook snout carp (Opsariichthys bidens). We found that the full-length cDNAs of O. bidens kif3a and kif3b were 2544 and 2806 bp in length comprising 119 bp and 259 bp 5' untranslated region (UTR), 313 bp and 222 bp 3' UTR, and 2112 bp and 2325 bp open reading frame encoding 703 and 774 amino acids, respectively. Ob-KIF3A/KIF3B proteins have three domains, namely N-terminal head, coiled-coil stalk, and C-terminal tail, and exhibit high similarity with homologous proteins in vertebrates and invertebrates. Ob-kif3a/kif3b mRNAs were ubiquitously expressed in all tissues examined, with the highest expression in the brain and stage-IV testis. Immunofluorescence results showed that Ob-KIF3A was co-localized with tubulin and the mitochondria. Particularly, in early spermatids, Ob-KIF3A, tubulin, and the mitochondrial signals were evenly distributed in the cytoplasm, whereas in middle spermatids, they were distributed around the nucleus. In the late stage, the signals were concentrated on one side of the nucleus, where the tail is formed, whereas in mature sperms, they were detected in the midpiece and flagellum. These results indicate that Ob-KIF3A/KIF3B may participate in nuclear reshaping, flagellum formation, and mitochondrial aggregation in the midpiece during spermiogenesis.


Subject(s)
Cyprinidae/physiology , Kinesins/physiology , Spermatogenesis/physiology , Animals , Cyprinidae/genetics , Kinesins/chemistry , Kinesins/genetics , Male , Microtubules/metabolism , Mitochondria/metabolism , Phylogeny , Protein Conformation , RNA, Messenger/metabolism , Sequence Alignment , Sequence Analysis, DNA , Sperm Tail/physiology , Spermatids/physiology , Spermatids/ultrastructure , Spermatogenesis/genetics , Testis/metabolism , Transcription, Genetic
5.
Genes (Basel) ; 12(5)2021 04 27.
Article in English | MEDLINE | ID: mdl-33925685

ABSTRACT

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


Subject(s)
Cell Nucleus/physiology , Cytoskeleton/metabolism , Cytoskeleton/physiology , Nuclear Envelope/metabolism , Nuclear Matrix/metabolism , Spermatozoa/metabolism , Spermatozoa/physiology , Animals , Cell Nucleus/metabolism , Humans , Infertility, Male/metabolism , Infertility, Male/physiopathology , Male , Mechanotransduction, Cellular/physiology , Nuclear Matrix/physiology , Spermatids/metabolism , Spermatids/physiology , Spermatocytes/metabolism , Spermatocytes/physiology
6.
Mol Reprod Dev ; 88(2): 141-157, 2021 02.
Article in English | MEDLINE | ID: mdl-33469999

ABSTRACT

BRDT, a member of the BET family of double bromodomain-containing proteins, is essential for spermatogenesis in the mouse and has been postulated to be a key regulator of transcription in meiotic and post-meiotic cells. To understand the function of BRDT in these processes, we first characterized the genome-wide distribution of the BRDT binding sites, in particular within gene units, by ChIP-Seq analysis of enriched fractions of pachytene spermatocytes and round spermatids. In both cell types, BRDT binding sites were mainly located in promoters, first exons, and introns of genes. BRDT binding sites in promoters overlapped with several histone modifications and histone variants associated with active transcription, and were enriched for consensus sequences for specific transcription factors, including MYB, RFX, ETS, and ELF1 in pachytene spermatocytes, and JunD, c-Jun, CRE, and RFX in round spermatids. Subsequent integration of the ChIP-seq data with available transcriptome data revealed that stage-specific gene expression programs are associated with BRDT binding to their gene promoters, with most of the BDRT-bound genes being upregulated. Gene Ontology analysis further identified unique sets of genes enriched in diverse biological processes essential for meiosis and spermiogenesis between the two cell types, suggesting distinct developmentally stage-specific functions for BRDT. Taken together, our data suggest that BRDT cooperates with different transcription factors at distinctive chromatin regions within gene units to regulate diverse downstream target genes that function in male meiosis and spermiogenesis.


Subject(s)
Epigenomics , Gene Expression Regulation, Developmental , Nuclear Proteins/physiology , Spermatogenesis/genetics , Transcription Factors/physiology , Animals , Binding Sites , Chromatin Immunoprecipitation Sequencing , DNA/metabolism , Male , Meiosis/genetics , Meiosis/physiology , Mice , Promoter Regions, Genetic , Spermatids/physiology , Spermatogenesis/physiology
7.
Asian J Androl ; 23(2): 123-128, 2021.
Article in English | MEDLINE | ID: mdl-32896837

ABSTRACT

Collagen α3 (IV) chains are one of the major constituent components of the basement membrane in the mammalian testis. Studies have shown that biologically active fragments, such as noncollagenase domain (NC1)-peptide, can be released from the C-terminal region of collagen α3 (IV) chains, possibly through the proteolytic action of metalloproteinase 9 (MMP9). NC1-peptide was shown to promote blood-testis barrier (BTB) remodeling and fully developed spermatid (e.g., sperm) release from the seminiferous epithelium because this bioactive peptide was capable of perturbing the organization of both actin- and microtubule (MT)-based cytoskeletons at the Sertoli cell-cell and also Sertoli-spermatid interface, the ultrastructure known as the basal ectoplasmic specialization (ES) and apical ES, respectively. More importantly, recent studies have shown that this NC1-peptide-induced effects on cytoskeletal organization in the testis are mediated through an activation of mammalian target of rapamycin complex 1/ribosomal protein S6/transforming retrovirus Akt1/2 protein (mTORC1/rpS6/Akt1/2) signaling cascade, involving an activation of cell division control protein 42 homolog (Cdc42) GTPase, but not Ras homolog family member A GTPase (RhoA), and the participation of end-binding protein 1 (EB1), a microtubule plus (+) end tracking protein (+TIP), downstream. Herein, we critically evaluate these findings, providing a critical discussion by which the basement membrane modulates spermatogenesis through one of its locally generated regulatory peptides in the testis.


Subject(s)
Basement Membrane/metabolism , Blood-Testis Barrier/metabolism , Collagen Type IV/metabolism , Peptide Fragments/metabolism , Seminiferous Epithelium/metabolism , Spermatogenesis/physiology , Actin Cytoskeleton , Animals , Basement Membrane/physiology , Blood-Testis Barrier/physiology , Cell Communication , Collagen Type IV/physiology , Humans , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules , Peptide Fragments/physiology , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6/metabolism , Seminiferous Epithelium/physiology , Sertoli Cells/metabolism , Sertoli Cells/physiology , Signal Transduction , Spermatids/metabolism , Spermatids/physiology , Testis , cdc42 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism
8.
Int J Mol Sci ; 21(22)2020 Nov 12.
Article in English | MEDLINE | ID: mdl-33198087

ABSTRACT

Male fertility disorders often have their origin in disturbed spermatogenesis, which can be induced by genetic factors. In this study, we used interspecific recombinant congenic mouse strains (IRCS) to identify genes responsible for male infertility. Using ultrasonography, in vivo and in vitro fertilization (IVF) and electron microscopy, the phenotyping of several IRCS carrying mouse chromosome 1 segments of Mus spretus origin revealed a decrease in the ability of sperm to fertilize. This teratozoospermia included the abnormal anchoring of the acrosome to the nucleus and a persistence of residual bodies at the level of epididymal sperm midpiece. We identified a quantitative trait locus (QTL) responsible for these phenotypes and we have proposed a short list of candidate genes specifically expressed in spermatids. The future functional validation of candidate genes should allow the identification of new genes and mechanisms involved in male infertility.


Subject(s)
Chromosomes, Human, Pair 1/genetics , Infertility, Male/genetics , Quantitative Trait Loci/genetics , Acrosome/physiology , Animals , Cell Nucleus/genetics , Cell Nucleus/physiology , Epididymis/physiology , Female , Humans , Male , Mice , Phenotype , Spermatids/physiology , Spermatogenesis/genetics , Spermatozoa/physiology , Teratozoospermia/genetics
9.
Reproduction ; 160(2): 319-330, 2020 08.
Article in English | MEDLINE | ID: mdl-32585638

ABSTRACT

Artificial oocyte activation is important for assisted reproductive technologies, such as fertilization with round spermatids (ROSI) or the production of cloned offspring by somatic cell nuclear transfer (SCNT). Recently, phospholipase Cζ (PLCζ)-cRNA was used to mimic the natural process of fertilization, but this method required the serial injection of PLCζ-cRNA and was found to cause damage to the manipulated oocytes. Here we tried to generate offspring derived from oocytes that were fertilized using round spermatid or somatic cell nuclear transfer with the co-injection of PLCζ-cRNA. After co-injecting round spermatids and 20 ng/µL of PLCζ-cRNA into the oocytes, most of them became activated, but the activation process was delayed by more than 1 h. With the co-injection method, the rate of blastocyst formation in ROSI embryos was higher (64%) compared with that of the serial injection method (55%). On another note, when SCNT was performed using the co-injection method, the cloned offspring were obtained with a higher success rate compared with the serial-injection method. However, in either ROSI or SCNT embryos, the birth rate of offspring via the co-injection method was similar to the Sr activation method. The epigenetic status of ROSI and SCNT zygotes that was examined showed no significant difference among all activation methods. The results indicated that although the PLCζ-cRNA co-injection method did not improve the production rate of offspring, this method simplified oocyte activation with less damage, and with accurate activation time in individual oocytes, it can be useful for the basic study of oocyte activation and development.


Subject(s)
Embryo, Mammalian/physiology , Nuclear Transfer Techniques/statistics & numerical data , Oocytes/physiology , Phosphoinositide Phospholipase C/metabolism , RNA, Complementary/administration & dosage , Spermatids/physiology , Zygote/physiology , Animals , Animals, Newborn , Embryo, Mammalian/cytology , Female , Male , Mice, Inbred ICR , Oocytes/cytology , Phosphoinositide Phospholipase C/administration & dosage , Phosphoinositide Phospholipase C/genetics , Pregnancy , Spermatids/cytology , Zygote/cytology
10.
Biol Reprod ; 103(3): 669-680, 2020 08 21.
Article in English | MEDLINE | ID: mdl-32406903

ABSTRACT

Tubulobulbar complexes (TBCs) internalize intercellular junctions during sperm release. One of the characteristic features of TBCs is that they form "bulbs" or swollen regions that have well-defined membrane contact sites (MCS) with adjacent cisternae of endoplasmic reticulum. Previously, we have localized the IP3R calcium channel to the TBC bulb-ER contacts and have hypothesized that fluctuations in local calcium levels may facilitate the maturation of TBC bulbs into putative endosomes, or alter local actin networks that cuff adjacent tubular regions of the TBCs. To test this, we injected the testes of Sprague Dawley rats with small interfering RNAs (siRNAs) against IP3R1 and processed the tissues for either western blot, immunofluorescence, or electron microscopy. When compared to control testes injected with nontargeting siRNAs, Sertoli cells in knocked-down testes showed significant morphological alterations to the actin networks including a loss of TBC actin and the appearance of ectopic para-crystalline actin bundles in Sertoli cell stalks. There also was a change in the abundance and distribution of TBC-ER contact sites and large internalized endosomes. This disruption of TBCs resulted in delay of the withdrawal of apical processes away from spermatids and in spermiation. Together, these findings are consistent with the hypothesis that calcium exchange at TBC-ER contacts is involved both in regulating actin dynamics at TBCs and in the maturing of TBC bulbs into endosomes. The results are also consistent with the hypothesis that TBCs are part of the sperm release mechanism.


Subject(s)
Inositol 1,4,5-Trisphosphate Receptors/genetics , Spermatids/ultrastructure , Testis/metabolism , Actins/biosynthesis , Actins/genetics , Animals , Calcium Signaling/genetics , Cell Communication , Gene Knockdown Techniques , Injections , Male , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Seminiferous Epithelium , Sertoli Cells , Spermatids/physiology , Spermatogenesis/genetics , Testis/cytology , Testis/ultrastructure
11.
Cell Rep ; 31(2): 107513, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32294451

ABSTRACT

Sertoli cells are supporting cells of the testicular seminiferous tubules, which provide a nurturing environment for spermatogenesis. Adult Sertoli cells are polarized so that they can simultaneously support earlier-stage spermatogenic cells (e.g., spermatogonia) basally and later-stage cells (e.g., spermatids) apically. To test the consequences of disrupting cell polarity in Sertoli cells, we perform a Sertoli-specific conditional deletion of Rac1, which encodes a Rho GTPase required for apicobasal cell polarity. Rac1 conditional knockout adults exhibit spermatogenic arrest at the round spermatid stage, with severe disruption of Sertoli cell polarity, and show increased germline and Sertoli cell apoptosis. Thus, Sertoli Rac1 function is critical for the progression of spermatogenesis but, surprisingly, is dispensable for fetal testicular development, adult maintenance of undifferentiated spermatogonia, and meiotic entry. Our data indicate that Sertoli Rac1 function is required only for certain aspects of spermatogenesis and reveal that there are distinct requirements for cell polarity during cellular differentiation.


Subject(s)
Neuropeptides/metabolism , Sertoli Cells/metabolism , Testis/cytology , rac1 GTP-Binding Protein/metabolism , Animals , Cell Differentiation/physiology , Cell Polarity/physiology , Male , Mice , Mice, Inbred C57BL , Neuropeptides/genetics , Seminiferous Tubules/cytology , Sertoli Cells/cytology , Sertoli Cells/pathology , Spermatids/physiology , Spermatogenesis/genetics , Spermatogenesis/physiology , Spermatogonia/physiology , Testis/growth & development , rac1 GTP-Binding Protein/genetics
12.
Zygote ; 28(4): 291-299, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32279700

ABSTRACT

Butterflyfish Chaetodon striatus is highly sought after in the marine ornamental aquarium, although studies about its reproductive biology are scarce. Therefore, to contribute to a better understanding of the reproductive aspects of C. striatus, we describe in detail with the use of high resolution histology the cellular dynamics of the germinal epithelium during the reproductive life history of this species. Based on the activity of the germinal epithelium, this study describes different stages of the gonadal development, similar to the reproductive phases found in other fish, to determine the reproductive period of C. striatus. In characterization of gonadal development, the following germ cells are described for males: spermatogonia, spermatocytes, spermatids and spermatozoa. Oogonia, early, primary, secondary, full-grown and maturing oocytes are described for females. Female germinal epithelium of C. striatus showed substantial changes over the study period, indicating that there was an active spawning period. Male germinal epithelium also presented relevant alterations, indicating reproductive activity in the testicular lobules. Morphological data confirm how informative was the cellular dynamics of the germinal epithelium for understanding gonadal development during adult reproductive life of fish in general. Although Chaetodon are a popular species, previous studies have only produced superficial and rough histological analyses. Therefore, this study demonstrates important information on germinal epithelium of Chaetodon. This knowledge could be a fundamental tool for development of new strategies for breeding of several species in captivity, especially butterflyfishes.


Subject(s)
Oocytes/growth & development , Ovary/growth & development , Perciformes/growth & development , Spermatozoa/growth & development , Testis/growth & development , Animals , Brazil , Epithelial Cells , Epithelium/metabolism , Female , Male , Oogenesis/physiology , Ovary/anatomy & histology , Ovary/cytology , Spermatids/physiology , Spermatocytes/physiology , Spermatogenesis/physiology , Testis/anatomy & histology , Testis/cytology
13.
PLoS One ; 15(4): e0230930, 2020.
Article in English | MEDLINE | ID: mdl-32267870

ABSTRACT

Human epidemiological studies have shown that paternal aging as one of the risk factors for neurodevelopmental disorders, such as autism, in offspring. A recent study has suggested that factors other than de novo mutations due to aging can influence the biology of offspring. Here, we focused on epigenetic alterations in sperm that can influence developmental programs in offspring. In this study, we qualitatively and semiquantitatively evaluated histone modification patterns in male germline cells throughout spermatogenesis based on immunostaining of testes taken from young (3 months old) and aged (12 months old) mice. Although localization patterns were not obviously changed between young and aged testes, some histone modification showed differences in their intensity. Among histone modifications that repress gene expression, histone H3 lysine 9 trimethylation (H3K9me3) was decreased in the male germline cells of the aged testis, while H3K27me2/3 was increased. The intensity of H3K27 acetylation (ac), an active mark, was lower/higher depending on the stages in the aged testis. Interestingly, H3K27ac was detected on the putative sex chromosomes of round spermatids, while other chromosomes were occupied by a repressive mark, H3K27me3. Among other histone modifications that activate gene expression, H3K4me2 was drastically decreased in the male germline cells of the aged testis. In contrast, H3K79me3 was increased in M-phase spermatocytes, where it accumulates on the sex chromosomes. Therefore, aging induced alterations in the amount of histone modifications and in the differences of patterns for each modification. Moreover, histone modifications on the sex chromosomes and on other chromosomes seems to be differentially regulated by aging. These findings will help elucidate the epigenetic mechanisms underlying the influence of paternal aging on offspring development.


Subject(s)
Histones/genetics , Meiosis/genetics , Spermatocytes/physiology , Spermatogenesis/genetics , Testis/physiology , Acetylation , Animals , Epigenesis, Genetic/genetics , Epigenomics/methods , Gene Expression/genetics , Histone Code/genetics , Humans , Lysine/genetics , Male , Methylation , Mice , Protein Processing, Post-Translational/genetics , Sex Chromosomes/genetics , Spermatids/physiology
14.
Gene ; 731: 144335, 2020 Mar 20.
Article in English | MEDLINE | ID: mdl-31927007

ABSTRACT

Deleted in azoospermia-like (DAZL) is essential for mammalian spermatogenesis as it regulates proliferation, development, maturation and functional maintenance of male germ cells. Its expression and regulation vary with different species or in the same animal at different developmental stages, and despite its importance, very little is known about its roles in sheep, especially Tibetan sheep. To investigate the expression patterns and regulatory roles of DZAL in Tibetan sheep testis, testicular tissue was isolated from sheep at three crucial development stages: 3 months old, 1 year old and 3 years old. Using quantitative real-time PCR and Western blot, we found that DAZL mRNA first decreased and then increased with advancing age, while DAZL protein exhibited an opposite expression pattern, with first increased and subsequently decreased levels. Immunohistochemistry and immunofluorescence revealed that DAZL protein was located predominantly in the cytoplasm of Leydig cells and in both the cytoplasm and nucleus of spermatids. ELISA indicated that testosterone content within developing testes was first enhanced and then declined. Our results, taken together, demonstrate, for the first time, that DAZL gene is involved in Tibetan sheep spermatogenesis by regulating the development of spermatids in post-pubertal rams, along with a novel role in functional maintenance of Leydig cells in postnatal rams.


Subject(s)
RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Sheep , Testis/growth & development , Testis/metabolism , Animals , Female , Gene Expression Profiling/veterinary , Gene Expression Regulation, Developmental , Leydig Cells/physiology , Male , RNA-Binding Proteins/physiology , Sex Differentiation/genetics , Sexual Maturation/genetics , Sheep/genetics , Sheep/growth & development , Sheep/metabolism , Spermatids/physiology , Spermatogenesis/genetics , Testosterone/metabolism , Tibet , Tissue Distribution
15.
RNA Biol ; 17(3): 350-365, 2020 03.
Article in English | MEDLINE | ID: mdl-31869276

ABSTRACT

The discovery of a large number of long noncoding RNAs (lncRNAs), and the finding that they may play key roles in different biological processes, have started to provide a new perspective in the understanding of gene regulation. It has been shown that the testes express the highest amount of lncRNAs among different vertebrate tissues. However, although some studies have addressed the characterization of lncRNAs along spermatogenesis, an exhaustive analysis of the differential expression of lncRNAs at its different stages is still lacking. Here, we present the results for lncRNA transcriptome profiling along mouse spermatogenesis, employing highly pure flow sorted spermatogenic stage-specific cell populations, strand-specific RNAseq, and a combination of up-to-date bioinformatic pipelines for analysis. We found that the vast majority of testicular lncRNA genes are expressed at post-meiotic stages (i.e. spermiogenesis), which are characterized by extensive post-transcriptional regulation. LncRNAs at different spermatogenic stages shared common traits in terms of transcript length, exon number, and biotypes. Most lncRNAs were lincRNAs, followed by a high representation of antisense (AS) lncRNAs. Co-expression analyses showed a high correlation along the different spermatogenic stage transitions between the expression patterns of AS lncRNAs and their overlapping protein-coding genes, raising possible clues about lncRNA-related regulatory mechanisms. Interestingly, we observed the co-localization of an AS lncRNA and its host sense mRNA in the chromatoid body, a round spermatids-specific organelle that has been proposed as a reservoir of RNA-related regulatory machinery. An additional, intriguing observation is the almost complete lack of detectable expression for Y-linked testicular lncRNAs, despite that a high number of lncRNA genes are annotated for this chromosome.


Subject(s)
RNA, Long Noncoding/genetics , Spermatogenesis/physiology , Animals , Gene Expression Regulation , Male , Mice , RNA, Antisense , RNA, Messenger/metabolism , Reproducibility of Results , Spermatids/cytology , Spermatids/physiology , Spermatogenesis/genetics , Testis/cytology , Testis/physiology
16.
Development ; 146(21)2019 11 04.
Article in English | MEDLINE | ID: mdl-31597660

ABSTRACT

Retinoic acid (RA) is crucial for mammalian spermatogonia differentiation, and stimulates Stra8 expression, a gene required for meiosis. Certain fish species, including zebrafish, have lost the stra8 gene. While RA still seems important for spermatogenesis in fish, it is not known which stage(s) respond to RA or whether its effects are integrated into the endocrine regulation of spermatogenesis. In zebrafish, RA promoted spermatogonia differentiation, supported androgen-stimulated meiosis, and reduced spermatocyte and spermatid apoptosis. Follicle-stimulating hormone (Fsh) stimulated RA production. Expressing a dominant-negative RA receptor variant in germ cells clearly disturbed spermatogenesis but meiosis and spermiogenesis still took place, although sperm quality was low in 6-month-old adults. This condition also activated Leydig cells. Three months later, spermatogenesis apparently had recovered, but doubling of testis weight demonstrated hypertrophy, apoptosis/DNA damage among spermatids was high and sperm quality remained low. We conclude that RA signaling is important for zebrafish spermatogenesis but is not of crucial relevance. As Fsh stimulates androgen and RA production, germ cell-mediated, RA-dependent reduction of Leydig cell activity may form a hitherto unknown intratesticular negative-feedback loop.


Subject(s)
Androgens/physiology , Endocrine System/physiology , Follicle Stimulating Hormone/physiology , Signal Transduction , Spermatogenesis , Tretinoin/physiology , Animals , Busulfan/chemistry , Cell Differentiation/genetics , Feedback, Physiological , Gene Expression Regulation, Developmental , Male , Mice , Retinoids/physiology , Spermatids/physiology , Spermatocytes/physiology , Spermatogonia/physiology , Testis/physiology , Transgenes , Zebrafish
17.
Biol Reprod ; 101(4): 842-853, 2019 10 25.
Article in English | MEDLINE | ID: mdl-31290539

ABSTRACT

Enhancers are cis-elements that activate transcription and play critical roles in tissue- and cell type-specific gene expression. During spermatogenesis, genes coding for specialized sperm structures are expressed in a developmental stage- and cell type-specific manner, but the enhancers responsible for their expression have not been identified. Using the mouse acrosomal vesicle protein (Acrv1) gene that codes for the acrosomal protein SP-10 as a model, our previous studies have shown that Acrv1 proximal promoter activates transcription in spermatids; and the goal of the present study was to separate the enhancer responsible. Transgenic mice showed that three copies of the -186/-135 fragment (50 bp enhancer) placed upstream of the Acrv1 core promoter (-91/+28) activated reporter expression in testis but not somatic tissues (n = 4). Immunohistochemistry showed that enhancer activity was restricted to the round spermatids. The Acrv1 enhancer failed to activate transcription in the context of a heterologous core promoter (n = 4), indicating a likely requirement for enhancer-core promoter compatibility. Chromatin accessibility assays showed that the Acrv1 enhancer assumes a nucleosome-free state in male germ cells (but not liver), indicating occupancy by transcription factors. Southwestern assays (SWA) identified specific binding of the enhancer to a testis nuclear protein of 47 kDa (TNP47). TNP47 was predominantly nuclear and becomes abundant during the haploid phase of spermatogenesis. Two-dimensional SWA revealed the isoelectric point of TNP47 to be 5.2. Taken together, this study delineated a 50-bp enhancer of the Acrv1 gene for round spermatid-specific transcription and identified a putative cognate factor. The 50-bp enhancer could become useful for delivery of proteins into spermatids.


Subject(s)
Enhancer Elements, Genetic/physiology , Membrane Proteins/genetics , Spermatids/metabolism , Spermatogenesis/genetics , Animals , Male , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Organ Specificity/genetics , Spermatids/physiology , Transcription, Genetic/genetics
18.
Biol Reprod ; 100(6): 1673-1685, 2019 06 01.
Article in English | MEDLINE | ID: mdl-31087039

ABSTRACT

Family with sequence similarity 46, member C (FAM46C) is a highly conserved non-canonical RNA polyadenylation polymerase that is abundantly expressed in human and mouse testes and is frequently mutated in patients with multiple myeloma. However, its physiological role remains largely unknown. In this study, we found that FAM46C is specifically localized to the manchette of spermatids in mouse testes, a transient microtubule-based structure mainly involved in nuclear shaping and intra-flagellar protein traffic. Gene knockout of FAM46C in mice resulted in male sterility, characterized by the production of headless spermatozoa in testes. Sperm heads were intermittently found in the epididymides of FAM46C knockout mice, but their fertilization ability was severely compromised based on the results of intracytoplasmic sperm injection assays. Interestingly, our RNA-sequencing analyses of FAM46C knockout testes revealed that mRNA levels of only nine genes were significantly altered compared to wild-type ones (q < 0.05). When considering alternate activities for FAM46C, in vitro assays demonstrated that FAM46C does not exhibit protein kinase or AMPylation activity against general substrates. Together, our data show that FAM46C in spermatids is a novel component in fastening the sperm head and flagellum.


Subject(s)
Flagella/physiology , Polynucleotide Adenylyltransferase/physiology , Sperm Head/physiology , Spermatids/physiology , Spermatogenesis/genetics , Animals , Cell Differentiation/genetics , Cells, Cultured , Female , Flagella/metabolism , Infertility, Male/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Polynucleotide Adenylyltransferase/genetics , Pregnancy , Sperm Head/metabolism , Spermatids/cytology , Spermatozoa/physiology
19.
Hum Mol Genet ; 28(15): 2561-2572, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31009948

ABSTRACT

Gonadotropin-regulated testicular RNA helicase (GRTH/DDX25) is a testis specific member of the DEAD-box family of RNA helicases expressed in meiotic and haploid germ cells which plays an essential role in spermatogenesis. There are two species of GRTH the 56 kDa non-phospho and 61 kDa phospho forms. Our early studies revealed a missense mutation (R242H) of GRTH in azoospermic men that when expressed in COS1-cells lack the phospho-form of GRTH. To investigate the role of the phospho-GRTH species in spermatogenesis, we generated a GRTH knock-in (KI) transgenic mice with the R242H mutation. GRTH-KI mice are sterile with reduced testis size, lack sperm with spermatogenic arrest at round spermatid stage and loss of the cytoplasmic phospho-GRTH species. Electron microscopy studies revealed reduction in the size of chromatoid bodies (CB) of round spermatids (RS) and germ cell apoptosis. We observed absence of phospho-GRTH in the CB of RS. Complete loss of chromatin remodeling and related proteins such as TP2, PRM2, TSSK6 and marked reduction of their respective mRNAs and half-lives were observed in GRTH-KI mice. We showed that phospho-GRTH has a role in TP2 translation and revealed its occurrence in a 3' UTR dependent manner. These findings demonstrate the relevance of phospho-GRTH in the structure of the chromatoid body, spermatid development and completion of spermatogenesis and provide an avenue for the development of a male contraceptive.


Subject(s)
DEAD-box RNA Helicases/metabolism , Infertility, Male/genetics , Mutation, Missense , Protein Processing, Post-Translational , Spermatids/metabolism , Animals , Aspermia/genetics , Aspermia/metabolism , Aspermia/physiopathology , Chromatin Assembly and Disassembly , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/physiology , Gene Expression Regulation , Infertility, Male/metabolism , Infertility, Male/physiopathology , Male , Mice , Mice, Knockout , Phosphorylation , Protamines/genetics , Protein Serine-Threonine Kinases/genetics , Spermatids/pathology , Spermatids/physiology , Spermatogenesis
20.
J Reprod Dev ; 65(3): 245-250, 2019 Jun 14.
Article in English | MEDLINE | ID: mdl-30773507

ABSTRACT

Piezo-actuated intracytoplasmic sperm injection (Piezo-ICSI) is used as an efficient in vitro fertilization method with various animals. With this method, elongated spermatids are collected from testicular tissues and are easier to obtain from animals that unexpectedly die than ejaculate sperm. Additionally, elongated spermatid injection often results in the development of embryos and offspring. To develop assisted reproductive techniques (ARTs) for domestic cats, we examined the effects of oocyte activation on cleavage and embryo development after Piezo-ICSI with motile sperm (experiment 1) and after Piezo-ICSI with either testicular sperm or elongated spermatids (experiment 2). In experiment 1, the proportions of cleaved embryos, morulas, and blastocysts following Piezo-ICSI with ethanol activation were significantly higher (P < 0.05) than in the non-activated groups. However, the proportion of blastocysts and the blastocyst quality did not differ significantly (P > 0.05) between the ethanol-activated and non-activated groups. In experiment 2, the cleavage frequencies of oocytes after Piezo-ICSI of testicular sperm or elongated spermatids and ethanol activation were higher (P < 0.05) than that of oocytes in the non-activated group, but the occurrence of blastocyst formation and quality of blastocysts did not differ between the activated and non-activated groups. In summary, cat embryos can be produced by Piezo-actuated microinjection of elongated spermatids. Ethanol activation increased the frequency of cleavage, but it affected neither the occurrence of blastocyst development nor the quality of blastocysts. These results represent an expansion in the repertoire of ARTs that are potentially applicable to both domestic and endangered species of cats.


Subject(s)
Oocytes/cytology , Sperm Injections, Intracytoplasmic/veterinary , Spermatids/physiology , Spermatozoa/physiology , Animals , Blastocyst/cytology , Cats , Cleavage Stage, Ovum , Cryopreservation , Embryonic Development , Female , Fertilization in Vitro , Male , Microinjections , Ovary/cytology , Testis/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...