Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 517
Filter
Add more filters

Publication year range
1.
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
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.
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
4.
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
5.
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
6.
PLoS Genet ; 14(2): e1007233, 2018 02.
Article in English | MEDLINE | ID: mdl-29462142

ABSTRACT

The sex chromosomes are enriched with germline genes that are activated during the late stages of spermatogenesis. Due to meiotic sex chromosome inactivation (MSCI), these sex chromosome-linked genes must escape silencing for activation in spermatids, thereby ensuring their functions for male reproduction. RNF8, a DNA damage response protein, and SCML2, a germline-specific Polycomb protein, are two major, known regulators of this process. Here, we show that RNF8 and SCML2 cooperate to regulate ubiquitination during meiosis, an early step to establish active histone modifications for subsequent gene activation. Double mutants of Rnf8 and Scml2 revealed that RNF8-dependent monoubiquitination of histone H2A at Lysine 119 (H2AK119ub) is deubiquitinated by SCML2, demonstrating interplay between RNF8 and SCML2 in ubiquitin regulation. Additionally, we identify distinct functions of RNF8 and SCML2 in the regulation of ubiquitination: SCML2 deubiquitinates RNF8-independent H2AK119ub but does not deubiquitinate RNF8-dependent polyubiquitination. RNF8-dependent polyubiquitination is required for the establishment of H3K27 acetylation, a marker of active enhancers, while persistent H2AK119ub inhibits establishment of H3K27 acetylation. Following the deposition of H3K27 acetylation, H3K4 dimethylation is established as an active mark on poised promoters. Together, we propose a model whereby regulation of ubiquitin leads to the organization of poised enhancers and promoters during meiosis, which induce subsequent gene activation from the otherwise silent sex chromosomes in postmeiotic spermatids.


Subject(s)
Histones/metabolism , Polycomb-Group Proteins/physiology , Sex Chromosomes/genetics , Transcriptional Activation/genetics , Ubiquitin-Protein Ligases/physiology , Ubiquitination/genetics , Acetylation , Animals , Female , Male , Meiosis/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Sex Chromosomes/metabolism , Spermatids/physiology , Spermatogenesis/genetics
7.
Semin Cell Dev Biol ; 81: 71-77, 2018 09.
Article in English | MEDLINE | ID: mdl-28923514

ABSTRACT

In adult mammalian testes, spermatids, most notably step 17-19 spermatids in stage IV-VIII tubules, are aligned with their heads pointing toward the basement membrane and their tails toward the tubule lumen. On the other hand, these polarized spermatids also align across the plane of seminiferous epithelium, mimicking planar cell polarity (PCP) found in other hair cells in cochlea (inner ear). This orderly alignment of developing spermatids during spermiogenesis is important to support spermatogenesis, such that the maximal number of developing spermatids can be packed and supported by a fixed population of differentiated Sertoli cells in the limited space of the seminiferous epithelium in adult testes. In this review, we provide emerging evidence to demonstrate spermatid PCP in the seminiferous epithelium to support spermatogenesis. We also review findings in the field regarding the biology of spermatid cellular polarity (e.g., head-tail polarity and apico-basal polarity) and its inter-relationship to spermatid PCP. Furthermore, we also provide a hypothetical concept on the importance of PCP proteins in endocytic vesicle-mediated protein trafficking events to support spermatogenesis through protein endocytosis and recycling.


Subject(s)
Cell Polarity/physiology , Signal Transduction/physiology , Spermatids/physiology , Spermatogenesis/physiology , Animals , Humans , Male , Sertoli Cells/cytology , Sertoli Cells/metabolism , Spermatids/cytology , Testis/cytology , Testis/metabolism
8.
Semin Cell Dev Biol ; 81: 46-53, 2018 09.
Article in English | MEDLINE | ID: mdl-29174914

ABSTRACT

Non-receptor Src family kinases (SFKs), most notably c-Src and c-Yes, are recently shown to be expressed by Sertoli and/or germ cells in adult rat testes. Studies have shown that SFKs are involved in modulating the cell cytoskeletal function, and involved in endocytic vesicle-mediated protein endocytosis, transcytosis and/or recycling as well as intracellular protein degradation events. Furthermore, a knockdown to SFKs, in particular c-Yes, has shown to induce defects in spermatid polarity. These findings, coupled with emerging evidence in the field, thus prompt us to critically evaluate them to put forth a developing concept regarding the role of SFKs and cell polarity, which will become a basis to design experiments for future investigations.


Subject(s)
Cell Polarity/physiology , Sertoli Cells/metabolism , Testis/metabolism , src-Family Kinases/metabolism , Animals , Cytoskeleton/metabolism , Humans , Male , Sertoli Cells/cytology , Spermatids/cytology , Spermatids/physiology , Testis/cytology , src-Family Kinases/genetics
9.
Semin Cell Dev Biol ; 81: 88-96, 2018 09.
Article in English | MEDLINE | ID: mdl-29410206

ABSTRACT

It is conceivable that spermatid apico-basal polarity and spermatid planar cell polarity (PCP) are utmost important to support spermatogenesis. The orderly arrangement of developing germ cells in particular spermatids during spermiogenesis are essential to obtain structural and nutrient supports from the fixed number of Sertoli cells across the limited space of seminiferous epithelium in the tubules following Sertoli cell differentiation by ∼17 day postpartum (dpp) in rodents and ∼12 years of age at puberty in humans. Yet few studies are found in the literature to investigate the role of these proteins to support spermatogenesis. Herein, we briefly summarize recent findings in the field, in particular emerging evidence that supports the concept that apico-basal polarity and PCP are conferred by the corresponding polarity proteins through their effects on the actin- and microtubule (MT)-based cytoskeletons. While much research is needed to bridge our gaps of understanding cell polarity, cytoskeletal function, and signaling proteins, a critical evaluation of some latest findings as summarized herein provides some important and also thought-provoking concepts to design better functional experiments to address this important, yet largely expored, research topic.


Subject(s)
Actins/metabolism , Cell Polarity/physiology , Cytoskeleton/metabolism , Microtubules/metabolism , Spermatids/physiology , Animals , Humans , Male , Sertoli Cells/cytology , Sertoli Cells/metabolism , Testis/cytology , Testis/metabolism
10.
Development ; 144(14): 2683-2693, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28619825

ABSTRACT

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.


Subject(s)
Proteins/physiology , Spermatids/growth & development , Spermatids/physiology , Spermatogenesis/physiology , Animals , Carrier Proteins/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Cytoplasmic Dyneins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubules/physiology , Protein Transport/genetics , Protein Transport/physiology , Proteins/genetics , Sperm Tail/physiology , Sperm Tail/ultrastructure , Spermatids/cytology , Spermatogenesis/genetics
11.
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
12.
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
13.
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
14.
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
15.
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
16.
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
17.
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
18.
Hum Reprod ; 34(3): 403-413, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30753464

ABSTRACT

STUDY QUESTION: Can full spermatogenesis be achieved after xenotransplantation of prepubertal primate testis tissue to the mouse, in testis or subcutaneously? SUMMARY ANSWER: Intratesticular xenotransplantation supported the differentiation of immature germ cells from marmoset (Callithrix jacchus) into spermatids and spermatozoa at 4 and 9 months post-transplantation, while in subcutaneous transplants, spermatogenic arrest was observed at 4 months and none of the transplants survived at 9 months. WHAT IS KNOWN ALREADY: Auto-transplantation of cryopreserved immature testis tissue (ITT) could be a potential fertility restoration strategy for patients with complete loss of germ cells due to chemo- and/or radiotherapy at a young age. Before ITT transplantation can be used for clinical application, it is a prerequisite to demonstrate the feasibility of the technique and identify the conditions required for establishing spermatogenesis in primate ITT transplants. Although xenotransplantation of ITT from several species has resulted in complete spermatogenesis, in human and marmoset, ITT has not been successful. STUDY DESIGN, SIZE, DURATION: In this study, we used marmoset as a pre-clinical animal model. ITT was obtained from two 6-month-old co-twin marmosets. A total of 147 testis tissue pieces (~0.8-1.0 mm3 each) were transplanted into the testicular parenchyma (intratesticular; n = 40) or under the dorsal skin (ectopic; n = 107) of 4-week-old immunodeficient Swiss Nu/Nu mice (n = 20). Each mouse received one single marmoset testis tissue piece in each testis and 4-6 pieces subcutaneously. Xenotransplants were retrieved at 4 and 9 months post-transplantation and evaluations were performed with regards to transplant survival, spermatogonial quantity and germ cell differentiation. PARTICIPANTS/MATERIALS, SETTING, METHODS: Transplant survival was histologically evaluated by haematoxylin-periodic acid Schiff (H/PAS) staining. Spermatogonia were identified by MAGE-A4 via immunohistochemistry. Germ cell differentiation was assessed by morphological identification of different germ cell types on H/PAS stained sections. Meiotically active germ cells were identified by BOLL expression. CREM immunohistochemistry was performed to confirm the presence of post-meiotic germ cells and ACROSIN was used to determine the presence of round, elongating and elongated spermatids. MAIN RESULTS AND THE ROLE OF CHANCE: Four months post-transplantation, 50% of the intratesticular transplants and 21% of the ectopic transplants were recovered (P = 0.019). The number of spermatogonia per tubule did not show any variation. In 33% of the recovered intratesticular transplants, complete spermatogenesis was established. Overall, 78% of the intratesticular transplants showed post-meiotic differentiation (round spermatids, elongating/elongated spermatids and spermatozoa). However, during the same period, spermatocytes (early meiotic germ cells) were the most advanced germ cell type present in the ectopic transplants. Nine months post-transplantation, 50% of the intratesticular transplants survived, whilst none of the ectopic transplants was recovered (P < 0.0001). Transplants contained more spermatogonia per tubule (P = 0.018) than at 4 months. Complete spermatogenesis was observed in all recovered transplants (100%), indicating a progressive spermatogenic development in intratesticular transplants between the two time-points. Nine months post-transplantation, transplants contained more seminiferous tubules with post-meiotic germ cells (37 vs. 5%; P < 0.001) and fewer tubules without germ cells (2 vs. 8%; P = 0.014) compared to 4 months post-transplantation. LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: Although xenotransplantation of marmoset ITT was successful, it does not fully reflect all aspects of a future clinical setting. Furthermore, due to ethical restrictions, we were not able to prove the functionality of the spermatozoa produced in the marmoset transplants. WIDER IMPLICATIONS OF THE FINDINGS: In this pre-clinical study, we demonstrated that testicular parenchyma provides the required microenvironment for germ cell differentiation and long-term survival of immature marmoset testis tissue, likely due to the favourable temperature regulation, growth factors and hormonal support. These results encourage the design of new experiments on human ITT xenotransplantation and show that intratesticular transplantation is likely to be superior to ectopic transplantation for fertility restoration following gonadotoxic treatment in childhood. STUDY FUNDING/COMPETING INTEREST(S): This project was funded by the ITN Marie Curie Programme 'Growsperm' (EU-FP7-PEOPLE-2013-ITN 603568) and the scientific Fund Willy Gepts from the UZ Brussel (ADSI677). D.V.S. is a post-doctoral fellow of the Fonds Wetenschappelijk Onderzoek (FWO; 12M2815N). No conflict of interest is declared.


Subject(s)
Spermatogenesis , Testis/physiology , Testis/transplantation , Animals , Callithrix , Cell Differentiation , Cryopreservation , Germ Cells/cytology , Male , Mice , Seminiferous Tubules/physiology , Sertoli Cells/physiology , Spermatids/physiology , Spermatogonia/physiology , Spermatozoa/physiology , Transplantation, Heterologous
19.
Reproduction ; 157(1): 27-42, 2019 01.
Article in English | MEDLINE | ID: mdl-30394707

ABSTRACT

In nature, mammalian seasonal breeders undergo spermatogenetic arrest during the non-breeding season. In the large hairy armadillo Chaetophractus villosus, testis regression initiates with immature post-meiotic germ cells sloughing into the tubule lumen and continues with the death of the remaining spermatocytes. At the end of the regression period, only spermatogonia and Sertoli cells persist in the seminiferous epithelium. It has been suggested that cell sloughing is determined by changes in the adhesion complexes between Sertoli cells and spermatids, which are mediated by low intra-testicular testosterone levels. By immunofluorescence and Western blotting we studied key proteins of the N-cadherin/N-cadherin and A6B1-integrin/laminin interlocks that contribute to the complex Sertoli/spermatid adhesion system throughout the eight stages of the seminiferous epithelium cycle in the comparison between active and regressing testes. In active testis, B1-integrin, laminin G3, N-cadherin, B-catenin, P-B-catenin-Tyr654, FAK, P-FAK-Tyr397, SRC, P-SRC-Tyr416 proteins present a spermatogenetic cycle-dependent localisation pattern, unmaintained in regressing testes. In the latter, quantitative variations and changes in the phosphorylation state of protein FAK, SRC and B-catenin contribute to the disassembly of the N-cadherin/N-cadherin and A6B1-integrin/laminin interlocks, thus promoting the massive release of immature spermatids.


Subject(s)
Armadillos/physiology , Sertoli Cells/physiology , Spermatids/physiology , Testis/cytology , Testis/growth & development , Animals , Armadillos/growth & development , Cell Differentiation , Male , Organ Size , Seasons , Sexual Behavior, Animal/physiology , Spermatogenesis/physiology , Testis/physiology
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