Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 848, 2024 Jan 29.
Article in English | MEDLINE | ID: mdl-38287033

ABSTRACT

Male germ cell development requires precise regulation of gene activity in a cell-type and stage-specific manner, with perturbations in gene expression during spermatogenesis associated with infertility. Here, we use steady-state, nascent and single-cell RNA sequencing strategies to comprehensively characterize gene expression across male germ cell populations, to dissect the mechanisms of gene control and provide new insights towards therapy. We discover a requirement for pausing of RNA Polymerase II (Pol II) at the earliest stages of sperm differentiation to establish the landscape of gene activity across development. Accordingly, genetic knockout of the Pol II pause-inducing factor NELF in immature germ cells blocks differentiation to spermatids. Further, we uncover unanticipated roles for Pol II pausing in the regulation of meiosis during spermatogenesis, with the presence of paused Pol II associated with double-strand break (DSB) formation, and disruption of meiotic gene expression and DSB repair in germ cells lacking NELF.


Subject(s)
RNA Polymerase II , Semen , Male , Humans , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Semen/metabolism , Meiosis/genetics , Spermatogenesis/genetics , Gene Expression
2.
bioRxiv ; 2023 May 09.
Article in English | MEDLINE | ID: mdl-37215034

ABSTRACT

Male germ cell development requires precise regulation of gene activity in a cell-type and stage-specific manner, with perturbations in gene expression during spermatogenesis associated with infertility. Here, we use steady-state, nascent and single-cell RNA sequencing strategies to comprehensively characterize gene expression across male germ cell populations, to dissect the mechanisms of gene control and provide new insights towards therapy. We discover a requirement for pausing of RNA Polymerase II (Pol II) at the earliest stages of sperm differentiation to establish the landscape of gene activity across development. Accordingly, genetic knockout of the Pol II pause-inducing factor NELF in immature germ cells blocks differentiation to mature spermatids. Further, we uncover unanticipated roles for Pol II pausing in the regulation of meiosis during spermatogenesis, with the presence of paused Pol II associated with double strand break formation by SPO11, and disruption of SPO11 expression in germ cells lacking NELF.

3.
Andrology ; 11(5): 904-910, 2023 07.
Article in English | MEDLINE | ID: mdl-36793255

ABSTRACT

Precise spatiotemporal expression of cohorts of differentiation markers unique to spermatogonia, spermatocytes, and round spermatids punctuates spermatogenesis and ensures its completion. For example, genes coding for the synaptonemal complex or the acrosome or flagellum are expressed sequentially in a developmental stage- and germ cell-specific manner. But the transcriptional mechanisms governing the spatiotemporal order of gene expression within the seminiferous epithelium are poorly understood. Using the round spermatid-specific Acrv1 gene, which codes for the acrosomal protein SP-10 as a model, we learned that (1) the proximal promoter itself contains all the necessary cis-regulatory sequences, (2) an insulator prevents somatic cell expression of the testis-specific gene, (3) RNA II polymerase is loaded on the Acrv1 promoter but paused in spermatocytes, thus ensuring precise transcriptional elongation in round spermatids, and that (4) a transcriptional repressor binding protein of 43 kilodaltons (TDP-43) plays a role in maintaining the paused state in spermatocytes. Although the Acrv1 enhancer element has been narrowed down to 50 bp and its binding to a 47 kDa testis-abundant nuclear protein shown, the identity of the putative transcription factor responsible for activation of round spermatid-specific transcription remains elusive. Human male infertility is idiopathic with limited treatment options. Understanding transcriptional regulation of spermatogenesis has the potential to lead to future therapies for male infertility.


Subject(s)
Infertility, Male , Seminiferous Epithelium , Mice , Male , Humans , Animals , Seminiferous Epithelium/metabolism , Membrane Proteins/genetics , Spermatids/metabolism , Gene Expression Regulation , Spermatogenesis/genetics , Testis/metabolism , Infertility, Male/genetics , Gene Expression
4.
Biol Reprod ; 107(5): 1345-1359, 2022 11 14.
Article in English | MEDLINE | ID: mdl-35986894

ABSTRACT

TAR DNA binding protein of 43 kD (TDP-43) is an evolutionarily conserved, ubiquitously expressed transcription factor and RNA-binding protein with major human health relevance. TDP-43 is present in Sertoli and germ cells of the testis and is aberrantly expressed in the sperm of infertile men. Sertoli cells play a key role in spermatogenesis by offering physical and nutritional support to male germ cells. The current study investigated the requirement of TDP-43 in Sertoli cells. Conditional knockout (cKO) of TDP-43 in mouse Sertoli cells caused failure of spermatogenesis and male subfertility. The cKO mice showed decreased testis weight, and low sperm count. Testis showed loss of germ cell layers, presence of vacuoles, and sloughing of round spermatids, suggesting loss of contact with Sertoli cells. Using a biotin tracer, we found that the blood-testis barrier (BTB) was disrupted as early as postnatal day 24 and worsened in adult cKO mice. We noted aberrant expression of the junction proteins connexin-43 (gap junction) and N-cadherin (ectoplasmic specialization). Oil Red O staining showed a decrease in lipid droplets (phagocytic function) in tubule cross-sections, Sertoli cells cytoplasm, and in the lumen of seminiferous tubules of cKO mice. Finally, qRT-PCR showed upregulation of genes involved in the formation and/or maintenance of Sertoli cell junctions as well as in the phagocytic pathway. Sertoli cells require TDP-43 for germ cell attachment, formation and maintenance of BTB, and phagocytic function, thus indicating an essential role for TDP-43 in the maintenance of spermatogenesis.


Subject(s)
Semen , Sertoli Cells , Animals , Male , Mice , Blood-Testis Barrier , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Sertoli Cells/metabolism , Spermatids/metabolism , Spermatogenesis/genetics , Testis/metabolism
5.
J Biol Chem ; 297(5): 101231, 2021 11.
Article in English | MEDLINE | ID: mdl-34599968

ABSTRACT

Meiotic arrest is a common cause of human male infertility, but the causes of this arrest are poorly understood. Transactive response DNA-binding protein of 43 kDa (TDP-43) is highly expressed in spermatocytes in the preleptotene and pachytene stages of meiosis. TDP-43 is linked to several human neurodegenerative disorders wherein its nuclear clearance accompanied by cytoplasmic aggregates underlies neurodegeneration. Exploring the functional requirement for TDP-43 for spermatogenesis for the first time, we show here that conditional KO (cKO) of the Tardbp gene (encoding TDP-43) in male germ cells of mice leads to reduced testis size, depletion of germ cells, vacuole formation within the seminiferous epithelium, and reduced sperm production. Fertility trials also indicated severe subfertility. Spermatocytes of cKO mice showed failure to complete prophase I of meiosis with arrest at the midpachytene stage. Staining of synaptonemal complex protein 3 and γH2AX, markers of the meiotic synaptonemal complex and DNA damage, respectively, and super illumination microscopy revealed nonhomologous pairing and synapsis defects. Quantitative RT-PCR showed reduction in the expression of genes critical for prophase I of meiosis, including Spo11 (initiator of meiotic double-stranded breaks), Rec8 (meiotic recombination protein), and Rad21L (RAD21-like, cohesin complex component), as well as those involved in the retinoic acid pathway critical for entry into meiosis. RNA-Seq showed 1036 upregulated and 1638 downregulated genes (false discovery rate <0.05) in the Tardbp cKO testis, impacting meiosis pathways. Our work reveals a crucial role for TDP-43 in male meiosis and suggests that some forms of meiotic arrest seen in infertile men may result from the loss of function of TDP-43.


Subject(s)
DNA-Binding Proteins/deficiency , Gene Expression Regulation , Infertility, Male/metabolism , Meiotic Prophase I , Seminiferous Epithelium/metabolism , Spermatocytes/metabolism , Spermatogenesis , Animals , DNA-Binding Proteins/metabolism , Female , Infertility, Male/genetics , Male , Mice , Mice, Knockout
6.
Biochim Biophys Acta Mol Basis Dis ; 1867(4): 166063, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33412266

ABSTRACT

Spinal muscular atrophy (SMA) is one of the major genetic disorders associated with infant mortality. More than 90% of cases of SMA result from deletions of or mutations in the Survival Motor Neuron 1 (SMN1) gene. SMN2, a nearly identical copy of SMN1, does not compensate for the loss of SMN1 due to predominant skipping of exon 7. The spectrum of SMA is broad, ranging from prenatal death to infant mortality to survival into adulthood. All tissues, including brain, spinal cord, bone, skeletal muscle, heart, lung, liver, pancreas, gastrointestinal tract, kidney, spleen, ovary and testis, are directly and/or indirectly affected in SMA. Accumulating evidence on impaired mitochondrial biogenesis and defects in X chromosome-linked modifying factors, coupled with the sexual dimorphic nature of many tissues, point to sex-specific vulnerabilities in SMA. Here we review the role of sex in the pathogenesis of SMA.


Subject(s)
Muscular Atrophy, Spinal/pathology , Animals , Female , Humans , Infertility, Male/etiology , Infertility, Male/genetics , Infertility, Male/pathology , Male , Mitochondria/genetics , Mitochondria/pathology , Muscular Atrophy, Spinal/etiology , Muscular Atrophy, Spinal/genetics , Sex Factors , Survival of Motor Neuron 1 Protein/genetics , X Chromosome/genetics
7.
Mol Reprod Dev ; 87(8): 857-870, 2020 08.
Article in English | MEDLINE | ID: mdl-32743879

ABSTRACT

Sertoli cells play a vital role in spermatogenesis by offering physical and nutritional support to the differentiating male germ cells. They form the blood-testis barrier and secrete growth factors essential for germ cell differentiation. Sertoli cell primary cultures are critical for understanding the regulation of spermatogenesis; however, obtaining pure cultures has been a challenge. Rodent Sertoli cell isolation protocols do not rule out contamination by the interstitial or connective tissue cells. Sertoli cell-specific markers could be helpful, but there is no consensus. Vimentin, the most commonly used marker, is not specific for Sertoli cells since its expression has been reported in peritubular myoid cells, mesenchymal stem cells, fibroblasts, macrophages, and endothelial cells, which contaminate Sertoli cell preparations. Markers based on transcription and growth factors also have limitations. Thus, the impediment to obtaining pure Sertoli cell cultures pertains to both the method of isolation and marker usage. The aim of this review is to discuss improvements to current methods of rodent Sertoli cell primary cultures, assess the properties of prepubertal versus mature Sertoli cell cultures, and propose steps to improve cellular characterization. Potential benefits of using contemporary approaches, including lineage tracing, specific cell ablation, and RNA-seq for obtaining Sertoli-specific transcript markers are discussed. Evaluating the specificity and applicability of these markers at the protein level to characterize Sertoli cells in culture would be critical. This review is expected to positively impact future work using primary cultures of rodent Sertoli cells.


Subject(s)
Primary Cell Culture , Rodentia , Sertoli Cells/cytology , Animals , Cell Differentiation , Cells, Cultured , Humans , Male , Primary Cell Culture/methods , Primary Cell Culture/veterinary , Sertoli Cells/physiology , Spermatogenesis/physiology
8.
Mol Reprod Dev ; 87(8): 871-879, 2020 08.
Article in English | MEDLINE | ID: mdl-32735067

ABSTRACT

Sertoli cells play a key role in spermatogenesis by supporting the germ cells throughout differentiation. The isolation of Sertoli cells is essential to study their functions. However, the close contact of Sertoli cells with other testicular cell types and the high proliferation of contaminating cells are obstacles to obtain pure primary cultures. Current rodent Sertoli cell isolation protocols result in enriched, rather than pure Sertoli cells. Therefore, novel approaches are necessary to improve the purity of Sertoli cell primary cultures. The goal of this study is to obtain pure mouse Sertoli cells using lineage tracing and fluorescence-activated cell sorting (FACS). We bred the Amh-Cre mouse line with tdTomato line to generate mice constitutively expressing red fluorescence specifically in Sertoli cells. Primary cultures of Sertoli cells isolated from prepubertal mice showed that 79% of cells expressed tdTomato, as evaluated by fluorescence microscopy and flow cytometry; however, nearly all adherent cells were positive for vimentin. Most of the tomato-negative cells expressed α-smooth muscle actin (α-SMA), a peritubular myoid cell marker, but double-negative populations were also present. These findings suggest that vimentin lacks Sertoli cell-specificity and that α-SMA is not adequate to identify all of the contaminating cells. Upon FACS sorting; however, virtually 100% of the cells were tdTomato positive, expressed vimentin, but not α-SMA. Prepubertal mice yielded a higher number of Sertoli cells compared to adults, but both could be adequately sorted. In conclusion, our study shows that lineage tracing and sorting is an efficient strategy for acquiring pure populations of murine Sertoli cells.


Subject(s)
Cell Separation/methods , Cell Tracking/methods , Sertoli Cells/cytology , Animals , Cell Differentiation/physiology , Cell Lineage/physiology , Cell Separation/veterinary , Cell Tracking/veterinary , Flow Cytometry/methods , Fluorescence , Germ Cells/cytology , Male , Mice , Mice, Transgenic , Spermatogenesis/physiology , Testis/cytology
9.
Theriogenology ; 156: 214-221, 2020 Oct 15.
Article in English | MEDLINE | ID: mdl-32758798

ABSTRACT

The acrosome plays a critical role in sperm-oocyte interactions during fertilization. SP-10 is an acrosomal matrix protein, which is evolutionarily conserved among mammals. The SP-10 antibody has been shown to be useful for staging the seminiferous cycle in the mouse and human. A canonical acrosomal marker; however, has never been used for staging in the horse. The objectives of the present study were to investigate the presence of SP-10 within the horse acrosome using an anti-mouse SP-10 antibody, to classify spermatids based on the shape of the acrosome, and then to use that information to assign stages of the cycle of the seminiferous epithelium. Testes from mature stallions with history of normospermic ejaculates were used for immunohistochemistry. We found that the mouse SP-10 antibody stained the horse acrosome vividly in testis cross-sections, indicating evolutionary conservation. Previous methods based on morphology alone without the aid of an antibody marker showed 8 stages in the horse seminiferous epithelium. Morphological detail of the acrosome afforded by the SP-10 marker in this study identified 16 steps of spermatids. This, in turn, led to the identification of 12 distinct stages in the cycle of the seminiferous epithelium of the horse wherein stage I shows recently formed round spermatids and stage XII includes meiotic divisions; a classification that is consistent with other animal models. The SP-10 antibody marks the acrosome in a way that enables researchers in the field to identify stages of spermatogenesis in the horse easily. In conclusion, we demonstrated that immunolabeling for SP-10 can be an objective approach to stage the cycle of the seminiferous epithelium in normospermic stallions; future studies will determine if SP-10 could be used to assess testicular dysfunction.


Subject(s)
Seminiferous Epithelium , Spermatids , Acrosome , Animals , Horses , Male , Mice , Spermatogenesis , Testis
10.
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
11.
Mol Reprod Dev ; 84(8): 675-685, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28600885

ABSTRACT

TAR DNA-binding protein of 43 kDa (TDP-43) is an evolutionarily conserved, ubiquitously expressed, multi-functional DNA/RNA-binding protein with roles in gene transcription, mRNA splicing, stability, transport, micro RNA biogenesis, and suppression of transposons. Aberrant expression of TDP-43 in testis and sperm was recently shown to be associated with male infertility, which highlights the need to understand better the expression of TDP-43 in the testis. We previously cloned TDP-43 from a mouse testis cDNA library, and showed that it functions as a transcriptional repressor and regulates the precise spatiotemporal expression of the Acrv1 gene, which encodes the acrosomal protein SP-10, during spermatogenesis. Here, we performed immunoblotting and immunohistochemistry of the mouse testis using four separate antibodies recognizing the amino and carboxyl termini of TDP-43. TDP-43 is present in the nuclei of germ cells as well as Sertoli cells. TDP-43 expression begins in type B/intermediate spermatogonia, peaks in preleptotene spermatocytes, and becomes undetectable in leptotene and zygotene spermatocytes. Pachytene spermatocytes and early round spermatids again express TDP-43, but its abundance diminishes later in spermatids (at steps 5-8). Interestingly, two of the four antibodies showed TDP-43 expression in spermatids at steps 9-10, which coincides with the initial phase of the histone-to-protamine transition. Immunoreactivity patterns observed in the study suggest that TDP-43 assumes different conformational states at different stages of spermatogenesis. TDP-43 pathology has been extensively studied in the context of neurodegenerative diseases; its role in spermatogenesis warrants further detailed investigation of the involvement of TDP-43 in male infertility.


Subject(s)
DNA-Binding Proteins/analysis , DNA-Binding Proteins/metabolism , Seminiferous Epithelium/chemistry , Seminiferous Epithelium/metabolism , Spermatogenesis/physiology , Animals , Gene Expression Regulation/genetics , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL
12.
Biologics ; 11: 55-63, 2017.
Article in English | MEDLINE | ID: mdl-28496303

ABSTRACT

This article reviews briefly the making of an immunoprophylactic-cum-immunotherapeutic vaccine against leprosy. The vaccine is based on cultivable, heat-killed atypical mycobacteria, whose gene sequence is now known. It has been named Mycobacterium indicus pranii. It has received the approval of the Drug Controller General of India and the US Food and Drug Administration. Besides leprosy, M. indicus pranii has found utility in the treatment of category II ("difficult to treat") tuberculosis. It also heals ugly anogenital warts. It has preventive and therapeutic action against SP2/O myelomas. It is proving to be a potent adjuvant for enhancing antibody titers of a recombinant vaccine against human chorionic gonadotropin, with the potential of preventing pregnancy without derangement of ovulation and menstrual regularity in sexually active women.

13.
Semin Reprod Med ; 35(2): 167-172, 2017 03.
Article in English | MEDLINE | ID: mdl-28278534

ABSTRACT

TDP-43 (TAR DNA binding Protein of 43 kD) is a transcription factor and RNA-binding protein with diverse functions. We cloned TDP-43 from the mouse testis in a screen for promoter-binding proteins and showed that it functions as a transcriptional repressor. TDP-43 plays a role in maintaining the precise pattern of spatiotemporal expression of the spermatid-specific Acrv1 gene during spermatogenesis by facilitating RNA polymerase II pausing at the promoter. We also showed that TDP-43 plays a partial role in preventing somatic cell expression of the Acrv1 gene by acting as an insulator-binding protein. Since the discovery of a causative link to several neurodegenerative diseases 10 years ago, TDP-43 has emerged as a protein of major human health relevance. Aberrant posttranslational modifications, nuclear exit, and cytoplasmic aggregate formation contribute to loss of neuronal function in patients. Interestingly, aberrant TDP-43 expression has also been reported in the testis and sperm of infertile men. Finally, our unpublished work shows that TDP-43 is indispensable for sperm formation and male fertility. The potential role of TDP-43 in male germ cells and fertility is discussed in this review.


Subject(s)
DNA-Binding Proteins/metabolism , Fertility , Spermatogenesis , Testis/metabolism , Animals , DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Humans , Infertility, Male/genetics , Infertility, Male/metabolism , Infertility, Male/physiopathology , Male , Signal Transduction
14.
J Clin Invest ; 127(3): 1046-1060, 2017 Mar 01.
Article in English | MEDLINE | ID: mdl-28218625

ABSTRACT

Autoimmune responses to meiotic germ cell antigens (MGCA) that are expressed on sperm and testis occur in human infertility and after vasectomy. Many MGCA are also expressed as cancer/testis antigens (CTA) in human cancers, but the tolerance status of MGCA has not been investigated. MGCA are considered to be uniformly immunogenic and nontolerogenic, and the prevailing view posits that MGCA are sequestered behind the Sertoli cell barrier in seminiferous tubules. Here, we have shown that only some murine MGCA are sequestered. Nonsequestered MCGA (NS-MGCA) egressed from normal tubules, as evidenced by their ability to interact with systemically injected antibodies and form localized immune complexes outside the Sertoli cell barrier. NS-MGCA derived from cell fragments that were discarded by spermatids during spermiation. They egressed as cargo in residual bodies and maintained Treg-dependent physiological tolerance. In contrast, sequestered MGCA (S-MGCA) were undetectable in residual bodies and were nontolerogenic. Unlike postvasectomy autoantibodies, which have been shown to mainly target S-MGCA, autoantibodies produced by normal mice with transient Treg depletion that developed autoimmune orchitis exclusively targeted NS-MGCA. We conclude that spermiation, a physiological checkpoint in spermatogenesis, determines the egress and tolerogenicity of MGCA. Our findings will affect target antigen selection in testis and sperm autoimmunity and the immune responses to CTA in male cancer patients.


Subject(s)
Autoantigens/immunology , Immune Tolerance , Seminiferous Tubules/immunology , Spermatogenesis/immunology , Spermatozoa/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Humans , Male , Mice , Mice, Inbred BALB C , Sertoli Cells/immunology
15.
Reprod Fertil Dev ; 28(6): 713-22, 2016 Apr.
Article in English | MEDLINE | ID: mdl-25359468

ABSTRACT

Loss of function of TAR DNA-binding protein (TDP-43) has been implicated in neurodegenerative disorders in both humans and animal models. TDP-43 has also been shown to be cis-acting transcriptional repressor of the acrosome vesicle (Acrv) gene in mice. In the present study, we investigated the expression of the TDP-43 transcript (TARDBP) and protein in germ cells from 11 fertile and 98 subfertile men to verify its potential association with poor seminograms. The expression profile of TDP-43 was characterised in immature germ cells and spermatozoa from semen from fertile and subfertile men using reverse transcription-polymerase chain reaction, western blotting and immunofluorescence. Although germ cells from subfertile men tested negative for TARDBP, the full-length message of the same was detected in fertile men. TDP-43 was detected in spermatozoa from fertile men using western blot analysis and immunofluorescence. The expression of this protein was negligible in spermatozoa from men with primary spermatogenic dysfunction. We conclude that a deficiency in the TDP-43 expression is associated with defective spermatogenesis and male infertility. We propose that TDP-43 could be used as a marker of male factor infertility.


Subject(s)
DNA-Binding Proteins/metabolism , Down-Regulation , Gene Expression Regulation, Developmental , Infertility, Male/metabolism , Spermatogenesis , Spermatozoa/metabolism , Biomarkers/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Humans , India , Infertility, Male/pathology , Infertility, Male/physiopathology , Male , Microscopy, Confocal , Microscopy, Fluorescence , Molecular Weight , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Proteolysis , RNA Processing, Post-Transcriptional , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Semen Analysis , Severity of Illness Index , Spermatozoa/pathology , Young Adult
16.
Mol Reprod Dev ; 81(10): 896-907, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25158006

ABSTRACT

The study of spermatogenesis requires accurate identification of the stages of the cycle of the seminiferous epithelium. A stage refers to the unique association of germ cell types at a particular phase of development, as seen in a cross-sectioned seminiferous tubule. Stage-identification, however, is a daunting task. There are 12 stages represented in the mouse seminiferous epithelium. Stages are typically identified on the basis of the morphology of the developing acrosome of spermatids. Although the characteristic features of the acrosome are well-documented in ultrastructure images, a reagent that can highlight the subtle differences in acrosome shape under the light microscope is lacking. Here we demonstrate that a polyclonal antibody raised against the mouse acrosomal protein SP-10 is extremely useful for stage identification. Immunohistochemistry showed that the anti-SP-10 antibody is highly specific for the acrosome of spermatids, as no other cell type in the epithelium showed immunoreactivity. At lower magnification, the gross shape of the acrosome and the increasing intensity of immunostaining served as a guide for the identification of stages I-XII. At higher magnification, characteristic morphological features-such as whether the part of the acrosome that contacts the nuclear surface is round (stage III) or flat (stage IV) or curved (stage VI)-could be identified unambiguously. Overall, we present evidence that SP-10 is a useful marker for staging the cycle of the seminiferous epithelium. The anti-SP-10 antibody works well in different fixatives, on paraffin-embedded as well as cryosections, and has been shown to be useful for characterizing spermatogenic defects in mutant mice.


Subject(s)
Acrosome/metabolism , Cell Cycle/physiology , Membrane Proteins/metabolism , Seminiferous Epithelium/metabolism , Spermatids/metabolism , Spermatogenesis/physiology , Animals , Immunohistochemistry , Male , Membrane Proteins/genetics , Mice , Mice, Mutant Strains , Seminiferous Epithelium/cytology , Spermatids/cytology
17.
J Biol Chem ; 286(13): 10970-82, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21252238

ABSTRACT

TDP-43 is an evolutionarily conserved ubiquitously expressed DNA/RNA-binding protein. Although recent studies have shown its association with a variety of neurodegenerative disorders, the function of TDP-43 remains poorly understood. Here we address TDP-43 function using spermatogenesis as a model system. We previously showed that TDP-43 binds to the testis-specific mouse acrv1 gene promoter in vitro via two GTGTGT-motifs and that mutation of these motifs led to premature transcription in spermatocytes of an otherwise round spermatid-specific promoter. The present study tested the hypothesis that TDP-43 represses acrv1 gene transcription in spermatocytes. Plasmid chromatin immunoprecipitation demonstrated that TDP-43 binds to the acrv1 promoter through GTGTGT motifs in vivo. Reporter gene assays showed that TDP-43 represses acrv1 core promoter-driven transcription via the N-terminal RRM1 domain in a histone deacetylase-independent manner. Consistent with repressor role, ChIP on physiologically isolated germ cells confirmed that TDP-43 occupies the endogenous acrv1 promoter in spermatocytes. Surprisingly, however, TDP-43 remains at the promoter in round spermatids, which express acrv1 mRNA. We show that RNA binding-defective TDP-43, but not splice variant isoforms, relieve repressor function. Transitioning from repressive to active histone marks has little effect on TDP-43 occupancy. Finally, we found that RNA polymerase II is recruited but paused at the acrv1 promoter in spermatocytes. Because mutation of TDP-43 sites caused premature transcription in spermatocytes in vivo, TDP-43 may be involved in pausing RNAPII at the acrv1 promoter in spermatocytes. Overall, our study shows that TDP-43 is a transcriptional repressor and that it regulates spatiotemporal expression of the acrv1 gene during spermatogenesis.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation/physiology , Membrane Proteins/biosynthesis , Repressor Proteins/metabolism , Spermatids/metabolism , Spermatocytes/metabolism , Spermatogenesis/physiology , Testis/metabolism , Animals , DNA-Binding Proteins/genetics , Histones/genetics , Histones/metabolism , Male , Membrane Proteins/genetics , Mice , Mutation , Protein Structure, Tertiary , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Repressor Proteins/genetics , Response Elements/physiology , Spermatids/cytology , Spermatocytes/cytology , Testis/cytology , Transcription, Genetic/physiology
18.
J Androl ; 29(2): 186-97, 2008.
Article in English | MEDLINE | ID: mdl-17942973

ABSTRACT

Identification of transcription factors involved in the progression of spermatogenic cell differentiation is important for understanding the molecular mechanisms controlling spermatogenesis. To this end, we utilized the mouse SP-10 gene encoding a conserved acrosomal protein as an experimental model. Promoter analysis in transgenic mice had previously shown that the -186/-91 region of the SP-10 promoter was critical for spermatid-specific expression. Here, we focus on a purine (Pu) box (-agaaaa) located at -154, which is conserved in the mouse, monkey, and human SP-10 gene promoters. NF45 and NF90, which belong to the family of nuclear factor of activated T cells (NFAT), are known as Pu-box-binding proteins. We tested the potential of NF45 and NF90 to activate the SP-10 promoter via the Pu-box element. Immunohistochemistry showed the presence of NF45 and NF90 in the nuclei of pachytene spermatocytes, round spermatids, and Sertoli cells. In gel shift assays, recombinant NF45 bound to the mouse SP-10 promoter in an AGAAAA site-specific manner. Cotransfection of NF45 and NF90 up-regulated SP-10 promoter-driven luciferase expression in transiently transfected spermatogenic GC2 cell line; this up-regulation required the -AGAAAA- site. Furthermore, stimulation of the endogenous NF45-NF90 complex in Jurkat cells by phorbol myristate acetate + ionomycin up-regulated the SP-10 promoter activity in plasmid-based assays. In the context of chromatin, however, stimulation of NF45-NF90 alone was not sufficient to activate an SP-10 promoter-driven green fluorescent protein transgene. Based on these results, we propose that NF45 and NF90 have the potential to activate SP-10 gene transcription, and that a chromatin modification event must occur first in order to provide access to these transcription factors.


Subject(s)
Membrane Proteins/genetics , Nuclear Factor 45 Protein/physiology , Nuclear Factor 90 Proteins/physiology , Seminiferous Epithelium/metabolism , Animals , Base Sequence , Haplorhini , Humans , Male , Mice , Promoter Regions, Genetic/physiology , Sequence Alignment , Transcriptional Activation/physiology
19.
J Biol Chem ; 282(50): 36143-54, 2007 Dec 14.
Article in English | MEDLINE | ID: mdl-17932037

ABSTRACT

Regulation of cell type-specific gene transcription is central to cellular differentiation and development. During spermatogenesis, a number of testis-specific genes are expressed in a precise spatiotemporal order. How these genes remain silent in the somatic tissues is not well understood. Our previous studies using the round spermatid-specific mouse SP-10 gene, which codes for an acrosomal protein, revealed that its proximal promoter acts as an insulator and prevents expression in the somatic tissues. Here we report that the insulator tethers the SP-10 gene to the nuclear matrix in somatic tissues, sequestering the core promoter in the process, thus preventing transcription. In round spermatids where the SP-10 gene is expressed, this tethering is released. TAR DNA-binding protein of 43 kDa (TDP-43), previously shown to interact with the SP-10 insulator, was found to be in the 2 m NaCl-insoluble nuclear matrix fraction. TDP-43 prevented enhancer-promoter interactions when artificially recruited between the two by Gal4 strategy. Knockdown of TDP-43 using small interfering RNA released the enhancer-blocking effect of the SP-10 insulator in a stable cell culture model. Mutation of TDP-43 binding sites abolished this effect. Finally, a 50-bp subfragment of the SP-10 insulator, which includes TDP-43 binding sites, functioned as a minimal insulator in transgenic mice and silenced an otherwise ectopically expressed transgene in somatic tissues. The SP-10 insulator lacks CpG dinucleotides or CTCF binding sites. Thus, the present study characterized a novel vertebrate insulator in a physiological context and showed for the first time how a testis-specific gene is silenced in the somatic tissues by an insulator.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation/physiology , Insulator Elements/physiology , Membrane Proteins/biosynthesis , Spermatids/metabolism , Testis/metabolism , Animals , COS Cells , Chlorocebus aethiops , CpG Islands/genetics , DNA-Binding Proteins/genetics , Enhancer Elements, Genetic/physiology , Gene Silencing , Male , Membrane Proteins/genetics , Mice , Mice, Transgenic , Mutation , Nuclear Matrix/genetics , Nuclear Matrix/metabolism , Organ Specificity/physiology , Promoter Regions, Genetic/physiology , Protein Binding/physiology , Spermatids/cytology , Testis/cytology
20.
Ann N Y Acad Sci ; 1120: 95-103, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18184912

ABSTRACT

Testis-specific promoters are unique in that relatively short proximal promoters of several genes have been shown to be capable of directing tissue- and cell-type-specific expression in transgenic mice. How such small promoter fragments perform the dual functions of maintaining a silenced state in somatic tissues and activating gene expression in the correct germ-cell type in testis remains poorly understood. Studies from our laboratory using the round spermatid-specific SP-10 gene as an experimental model have provided some insights into the mechanisms involved. It was found that the proximal promoter of the SP-10 gene acts as a chromatin insulator or boundary element in somatic tissues and prevents transcription of the SP-10 gene. In round spermatids, the insulator function is relieved, thus facilitating the SP-10 gene transcription. Insulators act as enhancer blockers and/or barriers to heterochromatin to protect the programmed expression of a gene. Typically, insulators are separable from promoters. In the case of the SP-10 gene, however, the insulator overlaps the promoter and operates in a facultative manner. We hypothesize that the proximal promoters of some testis-specific genes have adapted the insulator function to maintain transcriptional silence in the somatic tissues.


Subject(s)
Insulator Elements/physiology , Testis/metabolism , Transcription, Genetic , Animals , Humans , Male , Membrane Proteins/genetics , Models, Biological , Models, Molecular , Organ Specificity/genetics , Promoter Regions, Genetic/physiology , Spermatids/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...