Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
FASEB J ; 34(2): 3105-3128, 2020 02.
Article in English | MEDLINE | ID: mdl-31909540

ABSTRACT

During the epithelial cycle of spermatogenesis, different sets of cellular events take place across the seminiferous epithelium in the testis. For instance, remodeling of the blood-testis barrier (BTB) that facilitates the transport of preleptotene spermatocytes across the immunological barrier and the release of sperms at spermiation take place at the opposite ends of the epithelium simultaneously at stage VIII of the epithelial cycle. These cellular events are tightly coordinated via locally produced regulatory biomolecules. Studies have shown that collagen α3 (IV) chains, a major constituent component of the basement membrane, release the non-collagenous (NC) 1 domain, a 28-kDa peptide, designated NC1-peptide, from the C-terminal region, via the action of MMP-9 (matrix metalloproteinase 9). NC1-peptide was found to be capable of inducing BTB remodeling and spermatid release across the epithelium. As such, the NC1-peptide is an endogenously produced biologically active peptide which coordinates these cellular events across the epithelium in stage VIII tubules. Herein, we used an animal model, wherein NC1-peptide cloned into the pCI-neo mammalian expression vector was overexpressed in the testis, to better understanding the molecular mechanism by which NC1-peptide regulated spermatogenic function. It was shown that NC1-peptide induced considerable downregulation on a number of cell polarity and planar cell polarity (PCP) proteins, and studies have shown these polarity and PCP proteins modulate spermatid polarity and adhesion via their effects on microtubule (MT) and F-actin cytoskeletal organization across the epithelium. More important, NC1-peptide exerted its effects by downregulating the expression of microtubule (MT) plus-end tracking protein (+TIP) called EB1 (end-binding protein 1). We cloned the full-length EB1 cDNA for its overexpression in the testis, which was found to block the NC1-peptide-mediated disruptive effects on cytoskeletal organization in Sertoli cell epithelium and pertinent Sertoli cell functions. These findings thus illustrate that NC1-peptide is working in concert with EB1 to support spermatogenesis.


Subject(s)
Actin Cytoskeleton/metabolism , Blood-Testis Barrier/metabolism , Collagen/pharmacology , Microtubule-Associated Proteins/metabolism , Peptides/pharmacology , Spermatids/metabolism , Spermatogenesis/drug effects , Animals , Blood-Testis Barrier/cytology , Collagen/chemistry , Male , Matrix Metalloproteinase 9/metabolism , Peptides/chemistry , Rats , Rats, Sprague-Dawley , Seminiferous Epithelium/cytology , Seminiferous Epithelium/metabolism , Spermatids/cytology , Tight Junctions/metabolism
2.
Andrologia ; 51(6): e13285, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31006889

ABSTRACT

The present study was designed to investigate the therapeutic effect of bone marrow MSC-derived factors on gonadotropic toxicity induced by busulfan in vivo. The conditioned media (CM) was obtained from MSCs in serum-free incubation for 48 hr and concentrated ~25-fold by ultrafiltration. The CM of HEK 293 cells was treated as control (293-CM). MSC-CM was injected into busulfan mice via caudal veins after 1 day of busulfan treatment for 2 weeks (200 µl per dose/twice weekly). Compared to the 293-CM group, testicular injury was delayed in MSC-CM group, including reduced vacuolations of cells in the basal compartment of the seminiferous epithelium and detachment of cells from basement membrane. Apoptotic spermatogenic cells were significantly decreased in MSC-CM group (p ï¼œ 0.05). Interesting N-cadherin,ICAM-1 and P-cadherin expressions significantly increased in MSC-CM group, while occludin, ZO-1 and connexin 43 expressions showed no difference among MSC-CM, 293-CM and busulfan groups. Present results suggest MSC-secreted factors protect spermatogenesis impairment after busulfan treatment by reducing the apoptosis of spermatogenic cells and enhancing intercellular adhesion molecule expressions.


Subject(s)
Blood-Testis Barrier/drug effects , Busulfan/toxicity , Culture Media, Conditioned/pharmacology , Infertility, Male/drug therapy , Mesenchymal Stem Cells/metabolism , Animals , Apoptosis/drug effects , Blood-Testis Barrier/cytology , Blood-Testis Barrier/pathology , Cadherins/metabolism , Cell Adhesion/drug effects , Culture Media, Conditioned/metabolism , Disease Models, Animal , HEK293 Cells , Humans , Infertility, Male/chemically induced , Infertility, Male/pathology , Intercellular Adhesion Molecule-1/metabolism , Male , Mice , Spermatogenesis/drug effects
3.
Development ; 145(13)2018 07 09.
Article in English | MEDLINE | ID: mdl-29899137

ABSTRACT

Spermatogenesis in mammals is a very complex, highly organized process, regulated in part by testosterone and retinoic acid (RA). Much is known about how RA and testosterone signaling pathways independently regulate this process, but there is almost no information regarding whether these two signaling pathways directly interact and whether RA is crucial for steroidogenic cell function. This study uses a transgenic mouse line that expresses a dominant-negative form of RA receptor α (RAR-DN) and the steroidogenic cell-specific Cre mouse line, Cyp17iCre, to generate male mice with steroidogenic cells unable to perform RA signaling. Testes of mutant mice displayed increased apoptosis of pachytene spermatocytes, an increased number of macrophages in the interstitium and a loss of advanced germ cells. Additionally, blocking RA signaling in Leydig cells resulted in increased permeability of the blood-testis barrier, decreased levels of the steroidogenic enzyme cytochrome P450 17a1 and decreased testosterone levels. Surprisingly, the epididymides of the mutant mice also displayed an abnormal phenotype. This study demonstrates that RA signaling is required in steroidogenic cells for their normal function and, thus, for male fertility.


Subject(s)
Blood-Testis Barrier/metabolism , Fertility/physiology , Retinoic Acid Receptor alpha/metabolism , Signal Transduction/physiology , Spermatocytes/metabolism , Spermatogenesis/physiology , Animals , Blood-Testis Barrier/cytology , Male , Mice , Mice, Transgenic , Retinoic Acid Receptor alpha/genetics , Spermatocytes/cytology , Steroid 17-alpha-Hydroxylase/genetics , Steroid 17-alpha-Hydroxylase/metabolism
4.
Protein Pept Lett ; 25(5): 440-445, 2018.
Article in English | MEDLINE | ID: mdl-29651939

ABSTRACT

BACKGROUND: Sertoli cell, over the past 30 years, have been elevated from simple mechanical elements to the rank of a "sentinel" in spermatogenesis. By delivering potent immunomodulatory and trophic proteins, Sertoli cells are unique cell type with a pivotal role in maintaining testis immune privilege and the immune-protection of the antigenic germ cells. CONCLUSIONS: The findings from SC transplantation studies utilizing experimental animal models of disease, demonstrate the presence of the same immuno-modulation properties and mechanisms at tissue and organ sites far from testis. The complex pathways that generate and maintain the immune tolerance involve the production of several immunomodulatory or immune-related proteins such as cytokines, chemokines, growth factors, mediators of the inflammation, complement inhibitors or adhesion molecules. A better definition and understanding of these Sertoli cell proteins and the mechanisms of immunoprotection should help to elucidate their role in the spermatogenic process. The demonstration of their capabilities in transplantation experiments suggests that Sertoli cells may be good candidates in cell therapy for a number of cell-mediated chronic diseases.


Subject(s)
Blood-Testis Barrier/immunology , Immune Tolerance , Sertoli Cells/immunology , Spermatogenesis/immunology , Animals , Blood-Testis Barrier/cytology , Humans , Male , Sertoli Cells/cytology
5.
Mol Hum Reprod ; 24(5): 244-259, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29518209

ABSTRACT

STUDY QUESTION: What is the physiological role of Rictor in spermatogenic cells? SUMMARY ANSWER: Germline expression of Rictor regulates spermatogonial differentiation and has an essential role in coordinating germ cells and Sertoli cells in maintaining intact cell-cell adhesion dynamics and cytoskeleton-based architecture in the seminiferous epithelium. WHAT IS KNOWN ALREADY: The mechanistic target of rapamycin (mTOR) resides in its functions as the catalytic subunits of the structurally and functionally distinct mTORC1 and mTORC2 complexes. In the mammalian testis, mTORC1 regulates spermatogonial stem cell self-renewal and differentiation, whereas mTORC2 is required for Sertoli cell function. In contrast to mTORC1, mTORC2 has been much less well studied. Rictor is a distinct component of the mTORC2 complex. STUDY DESIGN, SIZE, DURATION: We investigated the effects of germ cell-specific ablation of Rictor on testicular development by using a mouse model of germline-specific ablation of Rictor. PARTICIPANTS/MATERIALS, SETTING, METHODS: We analyzed the in-vivo functions of Rictor through different methods including histology, immunofluorescent staining, chromosome spreads, blood-testis barrier (BTB) integrity assays and RNA sequencing. MAIN RESULTS AND THE ROLE OF CHANCE: Mutant mice did not show a defect in meiotic synapsis or recombination, but exhibited compromised spermatogonial differentiation potential, disorganized cell-cell junctions, impaired BTB dynamics and defective spermiogenesis. Concomitantly, RNA-seq profiling revealed that many genes involved in adhesion and migration were expressed inappropriately. LARGE SCALE DATA: RNA-seq data are published in the SRA database (PRJNA419273). LIMITATIONS REASONS FOR CAUTION: A detailed analysis of the mechanisms underlying the phenotype needs further investigations. WIDER IMPLICATIONS OF THE FINDINGS: Our work provides previously unidentified in-vivo evidence that germline expression of Rictor plays a role in maintaining spermatogonial differentiation and cell-cell adhesion. These findings are important for understanding the regulation of spermatogenesis and have clinical implications for the effect of mTOR inhibitors on human fertility. STUDY FUNDING AND COMPETING INTEREST(S): This study was supported by National Key R&D Program of China (2016YFA0500902), National Natural Science Foundation of China (31471228 and 31771653), Jiangsu Science Foundation for Distinguished Young Scholars (BK20150047), and Natural Science Foundation of Jiangsu Province (BK20140897, 14KJA180005 and 14KJB310004) to K.Z. The authors declare no competing or financial interests.


Subject(s)
Cell Adhesion/physiology , Rapamycin-Insensitive Companion of mTOR Protein/metabolism , Spermatogenesis/physiology , Spermatogonia/metabolism , Testis/metabolism , Animals , Blood-Testis Barrier/cytology , Blood-Testis Barrier/metabolism , Germ Cells/cytology , Germ Cells/metabolism , Male , Mice , Rapamycin-Insensitive Companion of mTOR Protein/genetics , Sertoli Cells/cytology , Sertoli Cells/metabolism , Spermatogonia/cytology , Testis/cytology
6.
FASEB J ; 32(3): 1653-1664, 2018 03.
Article in English | MEDLINE | ID: mdl-29183964

ABSTRACT

The assembly of the blood-testis barrier (BTB) during postnatal development is crucial to support meiosis. However, the role of germ cells in BTB assembly remains unclear. Herein, KitW/KitWV mice were used as a study model. These mice were infertile, failing to establish a functional BTB to support meiosis due to c-Kit mutation. Transplantation of undifferentiated spermatogonia derived from normal mice into the testis of KitW/KitWV mice triggered functional BTB assembly, displaying cyclic remodeling during the epithelial cycle. Also, transplanted germ cells were capable of inducing Leydig cell testosterone production, which could enhance the expression of integral membrane protein claudin 3 in Sertoli cells. Early spermatocytes were shown to play a vital role in directing BTB assembly by expressing claudin 3, which likely created a transient adhesion structure to mediate BTB and cytoskeleton assembly in adjacent Sertoli cells. In summary, the positive modulation of germ cells on somatic cell function provides useful information regarding somatic-germ cell interactions.-Li, X.-Y., Zhang, Y., Wang, X.-X., Jin, C., Wang, Y.-Q., Sun, T.-C., Li, J., Tang, J.-X., Batool, A., Deng, S.-L., Chen, S.-R., Cheng, C. Y., Liu, Y.-X. Regulation of blood-testis barrier assembly in vivo by germ cells.


Subject(s)
Blood-Testis Barrier/metabolism , Claudin-3/biosynthesis , Leydig Cells/metabolism , Sertoli Cells/metabolism , Spermatogonia/metabolism , Animals , Blood-Testis Barrier/cytology , Claudin-3/genetics , Leydig Cells/cytology , Male , Mice , Mice, Transgenic , Sertoli Cells/cytology , Spermatogonia/cytology
7.
Results Probl Cell Differ ; 58: 225-51, 2016.
Article in English | MEDLINE | ID: mdl-27300181

ABSTRACT

A healthy man typically produces between 50 × 10(6) and 200 × 10(6) spermatozoa per day by spermatogenesis; in the absence of Sertoli cells in the male gonad, this individual would be infertile. In the adult testis, Sertoli cells are sustentacular cells that support germ cell development by secreting proteins and other important biomolecules that are essential for germ cell survival and maturation, establishing the blood-testis barrier, and facilitating spermatozoa detachment at spermiation. In the fetal testis, on the other hand, pre-Sertoli cells form the testis cords, the future seminiferous tubules. However, the role of pre-Sertoli cells in this process is much less clear than the function of Sertoli cells in the adult testis. Within this framework, we provide an overview of the biology of the fetal, pubertal, and adult Sertoli cell, highlighting relevant cell biology studies that have expanded our understanding of mammalian spermatogenesis.


Subject(s)
Sertoli Cells/cytology , Spermatogenesis , Spermatozoa/cytology , Testis/cytology , Animals , Blood-Testis Barrier/cytology , Blood-Testis Barrier/embryology , Blood-Testis Barrier/growth & development , Cell Differentiation , Humans , Male , Seminiferous Tubules/cytology , Seminiferous Tubules/embryology , Seminiferous Tubules/growth & development , Testis/embryology , Testis/growth & development
8.
Hum Reprod ; 31(4): 875-86, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26908839

ABSTRACT

STUDY QUESTION: Are Sertoli cell tight junctions (TJs) disrupted in men undergoing hormonal contraception? SUMMARY ANSWER: Localization of the key Sertoli cell TJ protein, claudin-11, was markedly disrupted by 8 weeks of gonadotropin suppression, the degree of which was related to the extent of adluminal germ cell suppression. WHAT IS KNOWN ALREADY: Sertoli cell TJs are vital components of the blood-testis barrier (BTB) that sequester developing adluminal meiotic germ cells and spermatids from the vascular compartment. Claudin-11 knockout mice are infertile; additionally claudin-11 is spatially disrupted in chronically gonadotropin-suppressed rats coincident with a loss of BTB function, and claudin-11 is disorganized in various human testicular disorders. These data support the Sertoli cell TJ as a potential site of hormonal contraceptive action. STUDY DESIGN, SIZE, DURATION: BTB proteins were assessed by immunohistochemistry (n = 16 samples) and mRNA (n = 18 samples) expression levels in available archived testis tissue from a previous study of 22 men who had undergone 8 weeks of gonadotropin suppression and for whom meiotic and post-meiotic germ cell numbers were available. The gonadotropin suppression regimens were (i) testosterone enanthate (TE) plus the GnRH antagonist, acyline (A); (ii) TE + the progestin, levonorgestrel, (LNG); (iii) TE + LNG + A or (iv) TE + LNG + the 5α-reductase inhibitor, dutasteride (D). A control group consisted of seven additional men, with three archived samples available for this study. PARTICIPANTS/MATERIALS, SETTINGS, METHODS: Immunohistochemical localization of claudin-11 (TJ) and other junctional type markers [ZO-1 (cytoplasmic plaque), ß-catenin (adherens junction), connexin-43 (gap junction), vinculin (ectoplasmic specialization) and ß-actin (cytoskeleton)] and quantitative PCR was conducted using matched frozen testis tissue. MAIN RESULTS AND THE ROLE OF CHANCE: Claudin-11 formed a continuous staining pattern at the BTB in control men. Regardless of gonadotropin suppression treatment, claudin-11 localization was markedly disrupted and was broadly associated with the extent of meiotic/post-meiotic germ cell suppression; claudin-11 staining was (i) punctate (i.e. 'spotty' appearance) at the basal aspect of tubules when the average numbers of adluminal germ cells were <15% of control, (ii) presented as short fragments with cytoplasmic extensions when numbers were 15-25% of control or (iii) remained continuous when numbers were >40% of control. Changes in localization of connexin-43 and vinculin were also observed (smaller effects than for claudin-11) but ZO-1, ß-catenin and ß-actin did not differ, compared with control. LIMITATIONS, REASONS FOR CAUTION: Claudin-11 was the only Sertoli cell TJ protein investigated, but it is considered to be the most pivotal of constituent proteins given its known implication in infertility and BTB function. We were limited to testis samples which had been gonadotropin-suppressed for 8 weeks, shorter than the 74-day spermatogenic wave, which may account for the heterogeneity in claudin-11 and germ cell response observed among the men. Longer suppression (12-24 weeks) is known to suppress germ cells further and claudin-11 disruption may be more uniform, although we could not access such samples. WIDER IMPLICATIONS OF THE FINDINGS: These findings are important for our understanding of the sites of action of male hormonal contraception, because they suggest that BTB function could be ablated following long-term hormone suppression treatment. STUDY FUNDING/COMPETING INTERESTS: National Health and Medical Research Council (Australia) Program Grants 241000 and 494802; Research Fellowship 1022327 (to R.I.M.) and the Victorian Government's Operational Infrastructure Support Program. None of the authors have any conflicts to disclose. TRIAL REGISTRATION NUMBER: Not applicable.


Subject(s)
Claudins/antagonists & inhibitors , Contraceptive Agents, Male/pharmacology , Down-Regulation/drug effects , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Sertoli Cells/drug effects , Tight Junctions/drug effects , 5-alpha Reductase Inhibitors/pharmacology , Adult , Androgens/pharmacology , Blood-Testis Barrier/cytology , Blood-Testis Barrier/drug effects , Blood-Testis Barrier/metabolism , Claudins/genetics , Claudins/metabolism , Dutasteride/pharmacology , Humans , Immunohistochemistry , Levonorgestrel/pharmacology , Male , Middle Aged , Oligopeptides/pharmacology , Protein Transport/drug effects , Reproducibility of Results , Sertoli Cells/cytology , Spermatogenesis/drug effects , Testosterone/analogs & derivatives , Testosterone/pharmacology , Young Adult
9.
Mol Cell Endocrinol ; 405: 1-13, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25666991

ABSTRACT

The interaction of ouabain with the sodium pump induces signalling cascades resembling those triggered by hormone/receptor interactions. In the rat Sertoli cell line 93RS2, ouabain at low concentrations stimulates the c-Src/c-Raf/Erk1/2 signalling cascade via its interaction with the α4 isoform of the sodium pump expressed in these cells, leading to the activation of the transcription factor CREB. As a result of this signalling sequence, ouabain stimulates expression of claudin-1 and claudin-11, which are also controlled by a CRE promoter. Both of these proteins are known to be essential constituents of tight junctions (TJ) between Sertoli cells, and as a result of the ouabain-induced signalling TJ formation between neighbouring Sertoli cells is significantly enhanced by the steroid. Thus, ouabain-treated cell monolayers display higher transepithelial resistance and reduced free diffusion of FITC-coupled dextran in tracer diffusion assays. Taking into consideration that the formation of TJ is indispensable for the maintenance of the blood-testis barrier (BTB) and therefore for male fertility, the actions of ouabain described here and the fact that this and other related cardiotonic steroids (CTS) are produced endogenously suggest a direct influence of ouabain/sodium pump interactions on the maintenance of the BTB and thereby an effect on male fertility. Since claudin-1 and claudin-11 are also present in other blood-tissue barriers, one can speculate that ouabain and perhaps other CTS influence the dynamics of these barriers as well.


Subject(s)
Cardiotonic Agents/pharmacology , Claudin-1/metabolism , Claudins/metabolism , Gene Expression/drug effects , Ouabain/pharmacology , Sertoli Cells/metabolism , Animals , Blood-Testis Barrier/cytology , Cell Line , Claudin-1/genetics , Claudins/genetics , Male , Rats , Sertoli Cells/drug effects , Tight Junctions/metabolism , Transcriptional Activation
10.
Am J Physiol Endocrinol Metab ; 307(9): E738-53, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25159326

ABSTRACT

In the testis, spermatids are polarized cells, with their heads pointing toward the basement membrane during maturation. This polarity is crucial to pack the maximal number of spermatids in the seminiferous epithelium so that millions of sperms can be produced daily. A loss of spermatid polarity is detected after rodents are exposed to toxicants (e.g., cadmium) or nonhormonal male contraceptives (e.g., adjudin), which is associated with a disruption on the expression and/or localization of polarity proteins. In the rat testis, fascin 1, an actin-bundling protein found in mammalian cells, was expressed by Sertoli and germ cells. Fascin 1 was a component of the ectoplasmic specialization (ES), a testis-specific anchoring junction known to confer spermatid adhesion and polarity. Its expression in the seminiferous epithelium was stage specific. Fascin 1 was localized to the basal ES at the Sertoli cell-cell interface of the blood-testis barrier in all stages of the epithelial cycle, except it diminished considerably at late stage VIII. Fascin 1 was highly expressed at the apical ES at stage VII-early stage VIII and restricted to the step 19 spermatids. Its knockdown by RNAi that silenced fascin 1 by ~70% in Sertoli cells cultured in vitro was found to perturb the tight junction-permeability barrier via a disruption of F-actin organization. Knockdown of fascin 1 in vivo by ~60-70% induced defects in spermatid polarity, which was mediated by a mislocalization and/or downregulation of actin-bundling proteins Eps8 and palladin, thereby impeding F-actin organization and disrupting spermatid polarity. In summary, these findings provide insightful information on spermatid polarity regulation.


Subject(s)
Actin Cytoskeleton/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Cytoplasm/metabolism , Microfilament Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , Spermatogenesis , Testis/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/ultrastructure , Animals , Blood-Testis Barrier/cytology , Blood-Testis Barrier/drug effects , Blood-Testis Barrier/metabolism , Blood-Testis Barrier/ultrastructure , Cell Polarity/drug effects , Cells, Cultured , Contraceptive Agents, Male/pharmacology , Cytoplasm/drug effects , Cytoplasm/ultrastructure , Gene Expression Regulation, Developmental/drug effects , Gene Silencing , Hydrazines/pharmacology , Indazoles/pharmacology , Male , Microfilament Proteins/antagonists & inhibitors , Microfilament Proteins/genetics , Protein Transport/drug effects , RNA, Small Interfering , Rats, Sprague-Dawley , Sertoli Cells/cytology , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Sertoli Cells/ultrastructure , Spermatids/cytology , Spermatids/drug effects , Spermatids/metabolism , Spermatids/ultrastructure , Spermatogenesis/drug effects , Testis/cytology , Testis/drug effects , Testis/ultrastructure , Tight Junctions/drug effects , Tight Junctions/metabolism , Tight Junctions/ultrastructure
11.
Semin Cell Dev Biol ; 30: 65-74, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24727349

ABSTRACT

Non-receptor protein tyrosine kinases are cytoplasmic kinases that activate proteins by phosphorylating tyrosine residues, which in turn affect multiple functions in eukaryotic cells. Herein, we focus on the role of non-receptor protein tyrosine kinases, most notably, FAK, c-Yes and c-Src, in the transport of spermatids across the seminiferous epithelium during spermatogenesis. Since spermatids, which are formed from spermatocytes via meiosis, are immotile haploid cells, they must be transported by Sertoli cells across the seminiferous epithelium during the epithelial cycle of spermatogenesis. Without the timely transport of spermatids across the epithelium, the release of sperms at spermiation fails to occur, leading to infertility. Thus, the molecular event pertinent to spermatid transport is crucial to spermatogenesis. We provide a critical discussion based on recent findings in this review. We also provide a hypothetical model on spermatid transport, and the role of non-receptor protein tyrosine kinases in this event. We also highlight areas of research that deserve attention by investigators in the field.


Subject(s)
Protein-Tyrosine Kinases/physiology , Sperm Transport , Spermatids/enzymology , Spermatogenesis , Animals , Blood-Testis Barrier/cytology , Blood-Testis Barrier/physiology , Humans , Male , Phosphorylation , Protein Processing, Post-Translational , Seminiferous Epithelium/cytology , Sertoli Cells/enzymology , Signal Transduction , Spermatids/physiology
12.
FASEB J ; 27(3): 1137-52, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23288930

ABSTRACT

In the mammalian testis, coexisting tight junctions (TJs), basal ectoplasmic specializations, and gap junctions (GJs), together with desmosomes near the basement membrane, constitute the blood-testis barrier (BTB). The most notable feature of the BTB, however, is the extensive network of actin filament bundles, which makes it one of the tightest blood-tissue barriers. The BTB undergoes restructuring to facilitate the transit of preleptotene spermatocytes at stage VIII-IX of the epithelial cycle. Thus, the F-actin network at the BTB undergoes cyclic reorganization via a yet-to-be explored mechanism. Rictor, the key component of mTORC2 that is known to regulate actin cytoskeleton, was shown to express stage-specifically at the BTB in the seminiferous epithelium. Its expression was down-regulated at the BTB in stage VIII-IX tubules, coinciding with BTB restructuring at these stages. Using an in vivo model, a down-regulation of rictor at the BTB was also detected during adjudin-induced BTB disruption, illustrating rictor expression is positively correlated with the status of the BTB integrity. Indeed, the knockdown of rictor by RNAi was found to perturb the Sertoli cell TJ-barrier function in vitro and the BTB integrity in vivo. This loss of barrier function was accompanied by changes in F-actin organization at the Sertoli cell BTB in vitro and in vivo, associated with a loss of interaction between actin and α-catenin or ZO-1. Rictor knockdown by RNAi was also found to impede Sertoli cell-cell GJ communication, disrupting protein distribution (e.g., occludin, ZO-1) at the BTB, illustrating that rictor is a crucial BTB regulator.


Subject(s)
Blood-Testis Barrier/metabolism , Carrier Proteins/biosynthesis , Cell Communication/physiology , Gap Junctions/metabolism , Multiprotein Complexes/metabolism , Stress Fibers/metabolism , TOR Serine-Threonine Kinases/metabolism , Actins/genetics , Actins/metabolism , Animals , Blood-Testis Barrier/cytology , Carrier Proteins/genetics , Cell Communication/drug effects , Down-Regulation/drug effects , Down-Regulation/genetics , Gap Junctions/genetics , Gene Knockdown Techniques , Hydrazines/pharmacology , Indazoles/pharmacology , Male , Mechanistic Target of Rapamycin Complex 2 , Multiprotein Complexes/genetics , Rapamycin-Insensitive Companion of mTOR Protein , Rats , Rats, Sprague-Dawley , Seminiferous Epithelium/cytology , Seminiferous Epithelium/metabolism , Sertoli Cells/cytology , Sertoli Cells/metabolism , Stress Fibers/genetics , TOR Serine-Threonine Kinases/genetics , Zonula Occludens-1 Protein/genetics , Zonula Occludens-1 Protein/metabolism
13.
Nat Commun ; 3: 1185, 2012.
Article in English | MEDLINE | ID: mdl-23149730

ABSTRACT

Cellular events that occur across the seminiferous epithelium in the mammalian testis during spermatogenesis are tightly coordinated by biologically active peptides released from laminin chains. Laminin-γ3 domain IV is released at the apical ectoplasmic specialization during spermiation and mediates restructuring of the blood-testis barrier, which facilitates the transit of preleptotene spermatocytes. Here we determine the biologically active domain in laminin-γ3 domain IV, which we designate F5 peptide, and show that the overexpression of this domain, or the use of a synthetic F5 peptide, in Sertoli cells with an established functional blood-testis barrier reversibly perturbs blood-testis barrier integrity in vitro and in the rat testis in vivo. This effect is mediated via changes in protein distribution at the Sertoli and Sertoli-germ-cell cell interface and by phosphorylation of focal adhesion kinase at Tyr(407). The consequences are perturbed organization of actin filaments in Sertoli cells, disruption of the blood-testis barrier and spermatid loss. The impairment of spermatogenesis suggests that this laminin peptide fragment may serve as a contraceptive in male rats.


Subject(s)
Laminin/chemistry , Peptides/pharmacology , Spermatogenesis/drug effects , Actins/metabolism , Amino Acid Sequence , Animals , Blood-Testis Barrier/cytology , Blood-Testis Barrier/drug effects , Blood-Testis Barrier/metabolism , Cell Adhesion/drug effects , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Male , Molecular Sequence Data , Mutation/genetics , Occludin/metabolism , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Peptides/chemistry , Phosphorylation/drug effects , Protein Structure, Tertiary , Protein Transport/drug effects , Rats , Rats, Sprague-Dawley , Sertoli Cells/cytology , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Spermatozoa/cytology , Spermatozoa/drug effects , Spermatozoa/metabolism , Tight Junctions/metabolism
14.
Proc Natl Acad Sci U S A ; 109(31): 12562-7, 2012 Jul 31.
Article in English | MEDLINE | ID: mdl-22797892

ABSTRACT

Focal adhesion kinase (FAK), a nonreceptor protein tyrosine kinase, displays phosphorylation-dependent localization in the seminiferous epithelium of adult rat testes. FAK is an integrated component of the blood-testis barrier (BTB) involved in regulating Sertoli cell adhesion via its effects on the occludin-zonula occludens-1 complex. Herein, we report that p-FAK-Tyr(407) and p-FAK-Tyr(397) display restricted spatiotemporal and almost mutually exclusive localization in the epithelium, affecting BTB dynamics antagonistically, with the former promoting and the latter disrupting the Sertoli cell tight junction-permeability barrier function. Using primary cultured Sertoli cells as an in vitro model that mimics the BTB in vivo both functionally and ultrastructurally, effects of FAK phosphorylation on BTB function were studied by expressing nonphosphorylatable and phosphomimetic mutants, with tyrosine replaced by phenylalanine (F) and glutamate (E), respectively. Compared with WT FAK, Y407E and Y397F mutations each promoted barrier function, and the promoting effect of the Y407E mutant was abolished in the Y397E-Y407E double mutant, demonstrating antagonism between Tyr(407) and Tyr(397). Furthermore, Y407E mutation induced the recruitment of actin-related protein 3 to the Sertoli cell-cell interface, where it became more tightly associated with neuronal Wiskott-Aldrich syndrome protein, promoting actin-related protein 2/3 complex activity. Conversely, Y407F mutation reduced the rate of actin polymerization at the Sertoli cell BTB. In summary, FAK-Tyr(407) phosphorylation promotes BTB integrity by strengthening the actin filament-based cytoskeleton. FAK serves as a bifunctional molecular "switch" to direct the cyclical disassembly and reassembly of the BTB during the epithelial cycle of spermatogenesis, depending on its phosphorylation status, to facilitate the transit of preleptotene spermatocytes across the BTB.


Subject(s)
Blood-Testis Barrier/enzymology , Focal Adhesion Kinase 1/metabolism , Sertoli Cells/metabolism , Tight Junctions/enzymology , Actins/genetics , Actins/metabolism , Amino Acid Substitution , Animals , Blood-Testis Barrier/cytology , Cytoskeleton/genetics , Cytoskeleton/metabolism , Focal Adhesion Kinase 1/genetics , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutation, Missense , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation/genetics , Protein Multimerization/physiology , Rats , Sertoli Cells/cytology , Spermatocytes/cytology , Spermatocytes/enzymology , Spermatogenesis/physiology , Tight Junctions/genetics , Wiskott-Aldrich Syndrome Protein/genetics , Wiskott-Aldrich Syndrome Protein/metabolism , Zonula Occludens-1 Protein
15.
Adv Exp Med Biol ; 763: 149-70, 2012.
Article in English | MEDLINE | ID: mdl-23397623

ABSTRACT

The Scribble polarity complex or module is one of the three polarity modules that regulate cell polarity in multiple epithelia including blood-tissue barriers. This protein complex is composed of Scribble, Lethal giant larvae (Lgl) and Discs large (Dlg), which are well conserved across species from fruitflies and worms to mammals. Originally identified in Drosophila and C. elegans where the Scribble complex was found to work with the Par-based and Crumbs-based polarity modules to regulate apicobasal polarity and asymmetry in cells and tissues during embryogenesis, their mammalian homologs have all been identified in recent years. Components of the Scribble complex are known to regulate multiple cellular functions besides cell polarity, which include cell proliferation, assembly and maintenance of adherens junction (AJ) and tight junction (TJ), and they are also tumor suppressors. Herein, we provide an update on the Scribble polarity complex and how this protein complex modulates cell adhesion with some emphasis on its role in Sertoli cell blood-testis barrier (BTB) function. It should be noted that this is a rapidly developing field, in particular the role of this protein module in blood-tissue barriers, and this short chapter attempts to provide the information necessary for investigators studying reproductive biology and blood-tissue barriers to design future studies. We also include results of recent studies from flies and worms since this information will be helpful in planning experiments for future functional studies in the testis to understand how Scribble-based proteins regulate BTB dynamics and spermatogenesis.


Subject(s)
Blood-Testis Barrier/metabolism , Cell Polarity , Drosophila Proteins/metabolism , Intestinal Mucosa/metabolism , Membrane Proteins/metabolism , Multiprotein Complexes/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Blood-Testis Barrier/cytology , Cell Adhesion , Cell Proliferation , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Gene Expression Regulation, Developmental , Humans , Immunohistochemistry , Intestinal Mucosa/cytology , Mammals/metabolism , Membrane Proteins/genetics , Multiprotein Complexes/genetics , Mutation , Protein Structure, Tertiary , Protein Transport , Signal Transduction , Spermatogenesis , Tight Junctions/genetics , Tight Junctions/metabolism , Tumor Suppressor Proteins/genetics
16.
Adv Exp Med Biol ; 763: 281-94, 2012.
Article in English | MEDLINE | ID: mdl-23397630

ABSTRACT

Spermatogenesis involves precise co-ordination of multiple cellular events that take place in the seminiferous epithelium composed of Sertoli cells and developing germ cells during the seminiferous epithelial cycle. Given the cyclic and co-ordinated nature of spermatogenesis, temporal and spatial expression of certain genes pertinent to a specific cellular event are essential. As such, transcriptional regulation is one of the major regulatory machineries in controlling the cell type- and stage-specific gene expression, some of which are under the influence of gonadotropins (e.g., FSH and LH) and sex steroids (e.g., testosterone and estradiol-17beta). Recent findings regarding transcriptional control of spermatogenesis, most notably target genes at the Sertoli-Sertoli and Sertoli-spermatid interface at the site of the blood-testis barrier (BTB) and apical ectoplasmic specialization (apical ES), respectively, involving in cell adhesion are reviewed and discussed herein. This is a much neglected area of research and a concerted effort by investigators is needed to understand transcriptional regulation of cell adhesion function in the testis particularly at the BTB during spermatogenesis.


Subject(s)
Blood-Testis Barrier/metabolism , Cell Adhesion , Spermatogenesis , Testis/metabolism , Transcription, Genetic , Animals , Blood-Testis Barrier/cytology , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Intercellular Junctions/genetics , Intercellular Junctions/metabolism , Male , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Seminiferous Epithelium/cytology , Seminiferous Epithelium/metabolism , Sertoli Cells/cytology , Sertoli Cells/metabolism , Stem Cell Niche , Testis/cytology , Transcription Factors/genetics , Transcription Factors/metabolism
17.
Adv Exp Med Biol ; 763: 295-317, 2012.
Article in English | MEDLINE | ID: mdl-23397631

ABSTRACT

Src family kinases (SFKs), in particular c-Src and c-Yes, are nonreceptor protein tyrosine kinases that mediate integrin signaling at focal adhesion complex at the cell-extracellular matrix interface to regulate cell adhesion, cell cycle progression, cell survival, proliferation and differentiation, most notably in cancer cells during tumorigenesis and metastasis. Interestingly, recent studies have shown that these two proto-oncogenes are integrated components of the stem cell niche and the cell-cell actin-based anchoring junction known as ectoplasmic specialization (ES) at the: (1) Sertoli cell-spermatid interface known as apical ES and (2) Sertoli-Sertoli cell interface known as basal ES which together with tight junctions (TJ), gap junctions and desmosomes constitute the blood-testis barrier (BTB). At the stem cell niche, these SFKs regulate spermatogonial stem cell (SSC) renewal to maintain the proper population of SSC/spermatogonia for spermatogenesis. At the apical ES and the BTB, c-Src and c-Yes confer cell adhesion either by maintaining the proper phosphorylation status of integral membrane proteins at the site which in turn regulates protein-protein interactions between integral membrane proteins and their adaptors, or by facilitating androgen action on spermatogenesis via a nongenomic pathway which also modulates cell adhesion in the seminiferous epithelium. Herein, we critically evaluate recent findings in the field regarding the roles of these two unlikely partners of spermatogenesis. We also propose a hypothetical model on the mechanistic functions of c-Src and c-Yes in spermatogenesis so that functional experiments can be designed in future studies.


Subject(s)
Blood-Testis Barrier/enzymology , Proto-Oncogene Proteins c-yes/metabolism , Spermatogenesis , src-Family Kinases/metabolism , Animals , Apoptosis , Blood-Testis Barrier/cytology , CSK Tyrosine-Protein Kinase , Cell Adhesion , Cell Movement , Cell Proliferation , Enzyme Activation , Fertility , Focal Adhesions/enzymology , Focal Adhesions/genetics , Focal Adhesions/metabolism , Humans , Male , Membrane Proteins/metabolism , Models, Biological , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Phosphorylation , Protein Interaction Mapping , Proto-Oncogene Proteins c-yes/genetics , Seminiferous Epithelium/enzymology , Sertoli Cells/cytology , Sertoli Cells/metabolism , Stem Cell Niche , Testis/cytology , Testis/enzymology , src-Family Kinases/genetics
18.
Cell Stem Cell ; 9(5): 463-75, 2011 Nov 04.
Article in English | MEDLINE | ID: mdl-22056142

ABSTRACT

The homing ability of spermatogonial stem cells (SSCs) allows them to migrate into niches after being transplantated into infertile testes. Transplanted SSCs attach to Sertoli cells and transmigrate through the blood-testis barrier (BTB), formed by inter-Sertoli tight junctions, toward niches on the basement membrane. The most critical step is the passage through the BTB, which limits the homing efficiency to <10%. Here we demonstrated the involvement of Rac1 in SSC transmigration. Rac1-deficient SSCs did not colonize the adult testes, but they reinitiated spermatogenesis when transplanted into pup testes without a BTB. Moreover, a dominant-negative Rac1 construct not only reduced the expression of several claudin proteins, which comprise the BTB, but also increased SSC proliferation both in vitro and in vivo. Short hairpin RNA (shRNA) -mediated suppression of claudin3, which was downregulated by Rac inhibition, reduced the SSC homing efficiency. Thus, Rac1 is a critical regulator of SSC homing and proliferation.


Subject(s)
Blood-Testis Barrier/metabolism , Neuropeptides/metabolism , Spermatogonia/cytology , Stem Cell Niche , Stem Cells/cytology , Transendothelial and Transepithelial Migration , rac GTP-Binding Proteins/metabolism , Animals , Blood-Testis Barrier/cytology , Cell Shape , Cells, Cultured , Colony-Forming Units Assay , Embryo, Mammalian/cytology , Enzyme Activation , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Deletion , Gene Expression Regulation , Male , Mice , Mice, Knockout , Mutation/genetics , Neuropeptides/genetics , Spermatogonia/metabolism , Spermatogonia/transplantation , Stem Cells/metabolism , Tight Junctions/metabolism , rac GTP-Binding Proteins/genetics , rac1 GTP-Binding Protein
19.
Methods Mol Biol ; 763: 237-52, 2011.
Article in English | MEDLINE | ID: mdl-21874456

ABSTRACT

The use of an in vitro system based on primary cultures of Sertoli cells isolated from rat testes has greatly facilitated the study of the blood-testis barrier in recent years. Herein, we summarize the detailed procedures on the isolation of undifferentiated Sertoli cells from 20-day-old rat testes, the culture of these cells as a monolayer on Matrigel-coated bicameral units, the characterization of these cultured cells, and the use of the Sertoli cell epithelium for monitoring the integrity of the Sertoli cell blood-testis barrier. This information is based on the routine use of this system in our laboratory to study the Sertoli cell blood-testis barrier in the past two decades, which should be helpful for investigators in the field.


Subject(s)
Blood-Testis Barrier/metabolism , Cell Separation/methods , Potentiometry/methods , Sertoli Cells/physiology , Spermatogenesis/physiology , Tight Junctions/physiology , Animals , Animals, Newborn , Blood-Testis Barrier/cytology , Collagen/chemistry , Collagen/metabolism , Diffusion Chambers, Culture , Drug Combinations , Electric Impedance , Laminin/chemistry , Laminin/metabolism , Male , Microelectrodes , Microscopy, Fluorescence , Primary Cell Culture , Proteoglycans/chemistry , Proteoglycans/metabolism , Rats , Rats, Sprague-Dawley , Sertoli Cells/cytology
20.
Histochem Cell Biol ; 131(6): 755-64, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19241088

ABSTRACT

In mouse testis, claudin-11 is responsible for the formation of specific parallel TJ strands of the blood-testis barrier (BTB). Concerning the human BTB, there is no information about the transmembrane TJ proteins. We recently demonstrated the loss of functional integrity of the BTB in testicular intraepithelial neoplasia (TIN), associated with a dislocation of the peripheral TJ proteins ZO-1 and ZO-2. Here, we determined the expression and distribution of claudin-11 at the human BTB in seminiferous tubules with normal spermatogenesis (NSP) and TIN. Immunostaining of claudin-11 revealed intense signals at the basal BTB region in seminiferous epithelium with NSP. Within TIN tubules, claudin-11 immunostaining became diffuse and cytoplasmic. Double immunogold labeling demonstrated a co-localization of claudin-11 and ZO-1 at the inter-Sertoli cell junctions. Real-time RT-PCR of laser microdissected tubules showed an up-regulation of claudin-11 mRNA in TIN. Additionally, increased claudin-11 protein was observed by Western blot. We conclude that claudin-11 constitutes a TJ protein at the human BTB. In TIN tubules, claudin-11 is up-regulated and dislocated from the BTB. Therefore, the disruption of the BTB is related to a dysfunction of claudin-11 and not to a failure of its expression.


Subject(s)
Blood-Testis Barrier/metabolism , Nerve Tissue Proteins/metabolism , Seminiferous Epithelium/metabolism , Seminiferous Tubules/metabolism , Sertoli Cells/metabolism , Adult , Blood-Testis Barrier/cytology , Claudins , Humans , Male , Membrane Proteins/metabolism , Microscopy, Immunoelectron , Middle Aged , Phosphoproteins/metabolism , Seminiferous Epithelium/cytology , Seminiferous Epithelium/ultrastructure , Seminiferous Tubules/cytology , Seminiferous Tubules/ultrastructure , Sertoli Cells/cytology , Spermatogenesis/physiology , Testicular Neoplasms/metabolism , Testicular Neoplasms/ultrastructure , Testis/cytology , Testis/metabolism , Testis/ultrastructure , Up-Regulation , Zonula Occludens-1 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...