Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
1.
J Reprod Immunol ; 142: 103204, 2020 11.
Article in English | MEDLINE | ID: mdl-33130539

ABSTRACT

Detailed morphological characterization of testicular leukocytes in the adult CX3CR1 gfp/+ transgenic mouse identified two distinct CX3CR1 + mononuclear phagocyte (macrophage and dendritic cell) populations: stellate/dendriform cells opposed to the seminiferous tubules (peritubular), and polygonal cells associated with Leydig cells (interstitial). Using confocal microscopy combined with stereological enumeration of CX3CR1gfp/+ cells established that there were twice as many interstitial cells (68%) as peritubular cells (32%). Flow cytometric analyses of interstitial cells from mechanically-dissociated testes identified multiple mononuclear phagocyte subsets based on surface marker expression (CX3CR1, F4/80, CD11c). These cells comprised 80% of total intratesticular leukocytes, as identified by CD45 expression. The remaining leukocytes were CD3+ (T lymphocytes) and NK1.1+ (natural killer cells). Functional phenotype assessment using CD206 (an anti-inflammatory/M2 marker) and MHC class II (an activation marker) identified a potentially tolerogenic CD206+MHCII+ sub-population (12% of total CD45+ cells). Rare testicular subsets of CX3CR1 +CD11c+F4/80+ (4.3%) mononuclear phagocytes and CD3+NK1.1+ (3.1%) lymphocytes were also identified for the first time. In order to examine the potential for the immunoregulatory cytokine, activin A to modulate testicular immune cell populations, testes from adult mice with reduced activin A (Inhba+/-) or elevated activin A (Inha+/-) were assessed using flow cytometry. Although the proportion of F4/80+CD11b+ leukocytes (macrophages) was not affected, the frequency of CD206+MHCII+cells was significantly lower and CD206+MHCII- correspondingly higher in Inha+/- testes. This shift in expression of MHCII in CD206+ macrophages indicates that changes in circulating and/or local activin A influence resident macrophage activation and phenotype and, therefore, the immunological environment of the testis.


Subject(s)
Activins/metabolism , Inhibin-beta Subunits/metabolism , Leukocytes, Mononuclear/immunology , Macrophage Activation , Testis/immunology , Activins/analysis , Activins/genetics , Animals , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , Cell Separation , Flow Cytometry , Inhibin-beta Subunits/analysis , Inhibin-beta Subunits/genetics , Leukocytes, Mononuclear/metabolism , Male , Mice , Mice, Transgenic , Testis/cytology
2.
Andrology ; 7(1): 31-41, 2019 01.
Article in English | MEDLINE | ID: mdl-30315637

ABSTRACT

BACKGROUND: Human testicular germ cell tumours (TGCT) arise from germ cell neoplasia in situ (GCNIS) cells that originate from foetal germ cell precursors. Activin A is central to normal foetal testis development, and its dysregulation may contribute to TGCT aetiology. OBJECTIVE: (i) To test whether the expression profiles of activin A targets in normal and neoplastic human testes indicates functional links with TGCT progression. (ii) To investigate whether activin A levels influence MMP activity in a neoplastic germ cell line. MATERIALS AND METHODS: (1) Bouin's fixed, paraffin-embedded human testes were utilized for PCR-based transcript analysis and immunohistochemistry. Samples (n = 5 per group) contained the following: (i) normal spermatogenesis, (ii) GCNIS or (iii) seminoma. CXCL12, CCL17, MMP2 and MMP9 were investigated. (2) The human seminoma-derived TCam-2 cell line was exposed to activin A (24 h), and target transcripts were measured by qRT-PCR (n = 4). ELISA (n = 4) and gelatin zymography (n = 3) showed changes in protein level and enzyme activity, respectively. RESULTS: (i) Cytoplasmic CXCL12 was detected in Sertoli and other somatic cells, including those surrounding seminoma cells. Anti-CCL17 labelled only the cytoplasm of Sertoli cells surrounding GCNIS, while anti-MMP2 and anti-MMP9 labelled germline and epithelial-like cells in normal and neoplastic testes. (ii) Exposing TCam-2 cells to activin A (50 ng/mL) elevated MMP2 and MMP9 transcripts (fourfold and 30-fold), while only MMP2 protein levels were significantly higher after activin A (5 ng/mL and 50 ng/mL) exposure. Importantly, gelatin zymography revealed activin A increased production of activated MMP2. DISCUSSION: Detection of CCL17 only in GCNIS tumours may reflect a change in Sertoli cell phenotype to a less mature state. Stimulation of MMP2 activity by activin A in TCam-2 cells suggests activin influences TGCT by modulating the tumour niche. CONCLUSION: This knowledge provides a basis for understanding how physiological changes that influence activin/TGF-ß superfamily signalling may alter germ cell fate.


Subject(s)
Activins/metabolism , Neoplasms, Germ Cell and Embryonal/pathology , Seminoma/pathology , Sertoli Cells/metabolism , Testicular Neoplasms/pathology , Activins/genetics , Adult , Chemokine CCL17/metabolism , Chemokine CXCL12/metabolism , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Middle Aged , RNA, Messenger/genetics , Testis/metabolism
3.
Andrology ; 6(2): 362-373, 2018 03.
Article in English | MEDLINE | ID: mdl-29381885

ABSTRACT

Snail transcription factors are key regulators of cellular transitions during embryonic development and tumorigenesis. The closely related SNAI1 and SNAI2 proteins induce epithelial-mesenchymal transitions (EMTs), acting predominantly as transcriptional repressors, while the functions of SNAI3 are unknown. An initial examination of Snai2-deficient mice provided evidence of deficient spermatogenesis. To address the hypothesis that Snail proteins are important for male fertility, this study provides the first comprehensive cellular expression profiles of all three mammalian Snail genes in the post-natal mouse testis. To evaluate Snail transcript expression profiles, droplet digital (dd) PCR and in situ hybridization were employed. Snai1, 2 and 3 transcripts are readily detected at 7, 14, 28 days post-partum (dpp) and 7 weeks (adult). Unique cellular expression was demonstrated for each by in situ hybridization and immunohistochemistry using Western blot-validated antibodies. SNAI1 and SNAI2 are in the nucleus of the most mature germ cell types at post-natal ages 10, 15 and 26. SNAI3 is only detected from 15 dpp onwards and is localized in the Sertoli cell cytoplasm. In the adult testis, Snai1 and Snai2 transcripts are detected in spermatogonia and spermatocytes, while Snai3 is in both germ and Sertoli cells. SNAI1 protein is evident in nuclei of spermatogonia, spermatocytes, round spermatids and elongated spermatids (Stages IX-XII). SNAI2 is present in the nuclei of spermatogonia and spermatocytes, with a faint signal detected in round spermatids. SNAI3 was detected only in Sertoli cell cytoplasm, as in juvenile testes. Additionally, colocalization of SNAI1 and SNAI2 with previously identified key binding partners, LSD1 and PRC2 complex components, provides strong evidence that these important functional interactions are conserved during spermatogenesis to control gene activity. These distinct expression profiles suggest that each Snail family member has unique functions during spermatogenesis.


Subject(s)
Snail Family Transcription Factors/genetics , Testis/metabolism , Animals , Fertility , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism , Spermatogenesis/physiology , Testis/growth & development , Transcriptome
4.
Andrology ; 5(4): 763-770, 2017 07.
Article in English | MEDLINE | ID: mdl-28544640

ABSTRACT

Testicular germ cell tumours (TGCT) typically contain high numbers of infiltrating immune cells, yet the functional nature and consequences of interactions between GCNIS (germ cell neoplasia in situ) or seminoma cells and immune cells remain unknown. A co-culture model using the seminoma-derived TCam-2 cell line and peripheral blood mononuclear cells (PBMC, n = 7 healthy donors) was established to investigate how tumour and immune cells each contribute to the cytokine microenvironment associated with TGCT. Three different co-culture approaches were employed: direct contact during culture to simulate in situ cellular interactions occurring within seminomas (n = 9); indirect contact using well inserts to mimic GCNIS, in which a basement membrane separates the neoplastic germ cells and immune cells (n = 3); and PBMC stimulation prior to direct contact during culture to overcome the potential lack of immune cell activation (n = 3). Transcript levels for key cytokines in PBMC and TCam-2 cell fractions were determined using RT-qPCR. TCam-2 cell fractions showed an immediate increase (within 24 h) in several cytokine mRNAs after direct contact with PBMC, whereas immune cell fractions did not. The high levels of interleukin-6 (IL6) mRNA and protein associated with TCam-2 cells implicate this cytokine as important to seminoma physiology. Use of PBMCs from different donors revealed a robust, repeatable pattern of changes in TCam-2 and PBMC cytokine mRNAs, independent of potential inter-donor variation in immune cell responsiveness. This in vitro model recapitulated previous data from clinical TGCT biopsies, revealing similar cytokine expression profiles and indicating its suitability for exploring the in vivo circumstances of TGCT. Despite the limitations of using a cell line to mimic in vivo events, these results indicate how neoplastic germ cells can directly shape the surrounding tumour microenvironment, including by influencing local immune responses. IL6 production by seminoma cells may be a practical target for early diagnosis and/or treatment of TGCT.


Subject(s)
Cell Communication , Germ Cells/metabolism , Interleukin-6/metabolism , Leukocytes, Mononuclear/metabolism , Seminoma/metabolism , Seminoma/pathology , Testicular Neoplasms/metabolism , Tumor Microenvironment , Cell Line, Tumor , Cell Survival , Coculture Techniques , Culture Media, Conditioned/metabolism , Germ Cells/pathology , Humans , Interleukin-6/genetics , Leukocytes, Mononuclear/pathology , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Seminoma/genetics , Testicular Neoplasms/genetics , Testicular Neoplasms/pathology
5.
Andrology ; 5(3): 578-588, 2017 05.
Article in English | MEDLINE | ID: mdl-28235253

ABSTRACT

Activin A is an important regulator of testicular and epididymal development and function, as well as inflammation and immunity. In the adult murine reproductive tract, activin A mRNA (Inhba) expression levels are highest in the caput epididymis and decrease progressively towards the distal vas deferens. The activin-binding protein, follistatin (FST), shows the opposite expression pattern, with exceptionally high levels of the Fst288 mRNA variant in the vas deferens. This unique pattern of expression suggests that activin A and follistatin, in particular FST288, play region-specific roles in regulating the epididymis and vas deferens. The cellular distribution of activin and follistatin and structural organization of the male reproductive tract was examined in wild-type and transgenic (TghFST315) mice lacking FST288. Compared to wild-type littermates, TghFST315 mice showed a 50% reduction in serum follistatin and a significant elevation of both activin A and B. Testicular, epididymal and seminal vesicle weights were reduced, but intra-testicular testosterone was normal. A decrease in the epididymal duct diameter in the corpus and thickening of the peritubular smooth muscle in the cauda, together with increased coiling of the proximal vas deferens, were observed in TghFST315 mice. No immune cell infiltrates were detected. Immunohistochemistry indicated that epithelial cells are the main source of activins and follistatin in the epididymis and vas deferens. Activin A, but not activin B, was also localized to sperm heads in the lumen of the epididymis and vas deferens. Expression of Inhba and another immunoregulatory gene, indoleamine-2,3-dioxygenase (Ido-1), was increased approximately twofold in the TghFST315 caput epididymis, but several other genes associated with immunoregulation, inflammation or fibrosis were unaffected. Our novel data indicate that disruption of follistatin expression has significant effects on the testis and epididymis, and suggest an association between activin A and indoleamine-2,3-dioxygenase in the caput epididymis, with implications for the epididymal immunoenvironment.


Subject(s)
Activins/metabolism , Follistatin/metabolism , Genitalia, Male/metabolism , Animals , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Male , Mice , Mice, Transgenic , Polymerase Chain Reaction
6.
Br J Cancer ; 110(10): 2604-14, 2014 May 13.
Article in English | MEDLINE | ID: mdl-24781282

ABSTRACT

BACKGROUND: Testicular germ cell tumours of young adults, seminoma or non-seminomas, are preceded by a pre-invasive precursor, carcinoma in situ (CIS), understood to arise through differentiation arrest of embryonic germ cells. Knowledge about the malignant transformation of germ cells is currently limited by the lack of experimental models. The aim of this study was to establish an experimental tissue culture model to maintain normal and malignant germ cells within their niche and allow investigation of treatment effects. METHODS: Human testis and testis cancer specimens from orchidectomies were cultured in 'hanging drops' and effects of activin A and follistatin treatment were investigated in seminoma cultures. RESULTS: Testis fragments with normal spermatogenesis or CIS cells were cultured for 14 days with sustained proliferation of germ cells and CIS cells and without increased apoptosis. Seminoma cultures survived 7 days, with proliferating cells detectable during the first 5 days. Activin A treatment significantly reduced KIT transcript and protein levels in seminoma cultures, thereby demonstrating a specific treatment response. CONCLUSIONS: Hanging drop cultures of human testis and testis cancer samples can be employed to delineate mechanisms governing growth of normal, CIS and tumorigenic germ cells retained within their niche.


Subject(s)
Activins/pharmacology , Cell Culture Techniques , Follistatin/pharmacology , Seminoma/pathology , Testicular Neoplasms/pathology , Testis/cytology , Adult , Antigens, Neoplasm/analysis , Apoptosis/drug effects , Carcinoma in Situ/pathology , Cells, Cultured , DNA Replication/drug effects , Gene Expression Profiling , Humans , Ki-67 Antigen/analysis , Male , Morphogenesis/drug effects , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Primary Cell Culture/methods , Proto-Oncogene Proteins c-kit/biosynthesis , Proto-Oncogene Proteins c-kit/genetics , Spermatogenesis/drug effects , Spermatozoa/drug effects , Transcription Factor AP-2/biosynthesis , Transcription Factor AP-2/genetics , Tumor Cells, Cultured
7.
Andrology ; 2(2): 267-74, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24574096

ABSTRACT

Although the contribution of Hedgehog (Hh) signalling to stem cell development and oncogenesis is well recognised, its importance for spermatogonial stem cells (SSCs) has not been established. Here we interrogate adult rat SSCs using an established model in which only undifferentiated spermatogonial cells remain in the testis at 15 weeks following irradiation, and spermatogonial differentiation is induced within 4 weeks by gonadotrophin-releasing hormone antagonist (GnRH-ant) administration. Synthesis of Hh pathway components in untreated adult rat testes was compared with that in irradiated testes prior to and after GnRH-ant exposure using in situ hybridization. In adult testes with complete spermatogenesis, the Desert Hedgehog ligand transcript, Dhh, was detected in Sertoli cells, some spermatogonia and in spermatocytes by in situ hybridization. Spermatogenic cells were identified as sites of Hh signalling through detection of transcripts encoding the Hh receptor, Ptc2 transcripts and proteins for the key downstream target of Hh signalling, Gli1 and the Hh transcriptional activator, Gli2. Remarkably, the undifferentiated spermatogonia present in irradiated adult rat testes contained Dhh in addition to Ptc2, Gli1 and Gli2, revealing the potential for an autocrine Hh signalling loop to sustain undifferentiated spermatogonial cells. These transcripts became undetectable by in situ hybridization following GnRH-ant induction of spermatogonial differentiation, however, detection of Gli1 protein in spermatogonia in all groups indicates that Hh signalling is sustained. This is the first evidence of active Hh signalling in mammalian male germline stem cells, as has been documented for some cancer stem cells.


Subject(s)
Adult Stem Cells/metabolism , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Hedgehog Proteins/metabolism , Spermatogonia/metabolism , Adult Stem Cells/radiation effects , Animals , Cell Differentiation/drug effects , Cell Differentiation/radiation effects , DNA-Binding Proteins/genetics , Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/pharmacology , Hedgehog Proteins/biosynthesis , Hormone Antagonists/pharmacology , Kruppel-Like Transcription Factors/genetics , Male , Patched Receptors , Promyelocytic Leukemia Zinc Finger Protein , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/genetics , Sertoli Cells/metabolism , Signal Transduction , Spermatocytes/metabolism , Spermatogenesis/physiology , Testis/cytology , Testis/metabolism , Testis/radiation effects , Zinc Finger Protein GLI1 , Zinc Finger Protein Gli2
8.
Andrology ; 1(3): 517-29, 2013 May.
Article in English | MEDLINE | ID: mdl-23495012

ABSTRACT

Seminoma and non-seminoma tumours increasingly occur within the western population. These tumours originate from carcinoma in situ (CIS) cells, which arise from dysfunctional gonocytes. CXCL12 and its receptors, CXCR4 and CXCR7, have been implicated in migration, proliferation and survival of gonocytes and their precursors and progeny, primordial germ cells and spermatogonial stem cells respectively. We previously found evidence that several miRNA molecules predicted to modulate CXCR4 signalling are differentially expressed during the differentiation of gonocytes into spermatogonia in mice. Bioinformatic analysis predicted these miRNA to modulate CXCR4 signalling, leading us to hypothesize that CXCL12-mediated CXCR4 signalling is involved in the disrupted differentiation of gonocytes that underpins CIS formation. Indeed, we detected CXCL12 in Sertoli cells of normal human testis, and relatively high expression in tumour stroma with concomitant weak staining in dispersed tumour cells. In contrast, CXCR4 was expressed in spermatogonial and meiotic germ cells of normal testis and in the majority of tumour cells. Quantitative RT-PCR identified elevated CXCR4 transcript levels in seminoma compared with normal testis and to non-seminoma, potentially reflecting the higher proportion of dysfunctional germ cells within seminomas. In the normal testis, expression of CXCR4 downstream signalling molecules phospho-MEK1/2 and phospho-ERK1/2 correlated with CXCR4/CXCL12 expression. Strikingly, this correlation was absent in seminoma and non-seminoma samples, suggesting that CXCL12 signalling is disrupted. Proliferation rate and cell survival were not altered by CXCL12 in either seminoma (TCam-2) or non-seminoma (833ke) cell lines. However, CXCL12 exposure induced TCam-2 cell invasion though simulated basement membrane, while in contrast, we provide the novel evidence that CXCR4-expressing non-seminoma cell lines 833ke and NTera2/D1 do not invade in response to CXCL12. These findings indicate that CXCL12 expression in the human testis may selectively influence seminoma migration and metastasis, correlating with its importance in gonocyte and spermatogonial stem cell biology.


Subject(s)
Chemokine CXCL12/physiology , Neoplasm Metastasis , Receptors, CXCR4/physiology , Seminoma/pathology , Cell Line, Tumor , Humans , Male
9.
Mol Cell Endocrinol ; 359(1-2): 66-77, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22406273

ABSTRACT

The discovery of activin and inhibins as modulators of the hypothalamic-pituitary-gonadal axis has set the foundation for understanding their central importance to many facets of development and disease. This review contains an overview of the processes and cell types that are central to testis development and spermatogenesis and then provides an update focussed on information gathered over the past five years to address new concepts about how these proteins function to control testis development in fetal and juvenile life. Current knowledge about the interactive nature of the transforming growth factor-ß (TGFß) superfamily signalling network is applied to recent findings about activins and inhibins in the testis. Information about the regulated synthesis of signalling components and signalling regulators in the testis is integrated with new concepts that demonstrate their functional significance. The importance of activin bioactivity levels or dosage in controlling balanced growth of spermatogonial cells and their niche at different stages of testis development is highlighted.


Subject(s)
Activins/metabolism , Inhibins/metabolism , Testis/embryology , Testis/growth & development , Activins/physiology , Animals , Humans , Inhibins/physiology , Male , Organogenesis , Signal Transduction , Spermatogenesis , TGF-beta Superfamily Proteins/metabolism , TGF-beta Superfamily Proteins/physiology , Testis/physiology
10.
Int J Androl ; 35(2): 158-69, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21812786

ABSTRACT

Spermatogenesis requires progressive changes in gene expression mediated by hormonal and local factors. Regulated macromolecular movement between nuclear and cytoplasmic compartments enables these essential responses to changing extracellular cues, and dynamic production of the nucleocytoplasmic transporters and importin proteins, throughout gametogenesis in rodents implicates them as key mediators of germline differentiation. We examined normal adult human testis expression profiles of six importins plus five additional proteins involved in nucleocytoplasmic transport. Although most were detected in the nucleus during germline differentiation, importin α4 was exclusively observed in Sertoli and germ cell cytoplasm. Many proteins were present in round spermatid nuclei (importins α1, α3, ß1, ß3; exportin-1, Nup62, Ran, RanBP1, RCC1), and remarkable intense nuclear and/or nuclear-associated signals were detected for importin α1, importin α3 and Nup62 in spermatocytes. This study identifies conserved aspects of nucleocytoplasmic transport during spermatogenesis and extends our knowledge of the dynamic presence of these proteins, which indicates that they contribute to germ cell-specific cargo trafficking and potentially to other functions during human spermatogenesis. We also demonstrate for the first time that importin α3 is nuclear in spermatocytes, when exportin-1 is cytoplasmic, suggesting that nuclear transport is altered during meiosis.


Subject(s)
Active Transport, Cell Nucleus , Nucleocytoplasmic Transport Proteins/metabolism , Spermatocytes/metabolism , Spermatogenesis , Animals , Cell Differentiation , Cell Line, Tumor , Cell Nucleus/metabolism , Cytoplasm/metabolism , Gene Expression , Gene Expression Regulation , Germ Cells/cytology , HeLa Cells , Humans , Karyopherins/biosynthesis , Karyopherins/metabolism , Male , Membrane Glycoproteins/biosynthesis , Mice , Mice, Inbred C57BL , Nuclear Pore Complex Proteins/biosynthesis , Receptors, Cytoplasmic and Nuclear/metabolism , Spermatids/metabolism , Exportin 1 Protein
11.
Int J Androl ; 34(4 Pt 2): e204-17, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21668453

ABSTRACT

Germ cell testicular cancer is understood to arise during embryogenesis, based on the persistence of embryonic germ cell markers in carcinoma in situ and seminoma. In this study, we examine the potential of the seminoma-derived TCam-2 cell line to be used as representative in functional analyses of seminoma. We demonstrate expression of several early germ cell markers, including BLIMP1, OCT3/4, AP2γ, NANOG and KIT. Many TGF-beta superfamily receptors and downstream transcription factors are also present in these cells including the normally foetal ACTRIIA receptor, indicating potential responsiveness to TGF-beta superfamily ligands. Treatment with BMP4 or RA induces a significant increase in ACTRIA, ACTRIIA and ACTRIIB transcripts, whereas activin A decreases ACTRIB. BMP4 and RA each support TCam-2 survival and/or proliferation. In addition, despite increased KIT mRNA levels induced by BMP4, RA and activin A, activin A does not improve survival or proliferation. The capacity for BMP4 and retinoic acid to enhance foetal germ cell survival and proliferation/self-renewal has been demonstrated in mice, but not previously tested in humans. This study is the first to demonstrate a functional response in seminoma cells, using a well-characterized cell line, consistent with their foetal germ cell-like identity.


Subject(s)
Germ Cells/drug effects , Seminoma/metabolism , Transforming Growth Factor beta/pharmacology , Tretinoin/pharmacology , Activin Receptors, Type II/metabolism , Activins/pharmacology , Adaptor Protein Complex 2/biosynthesis , Biomarkers , Bone Morphogenetic Protein 4 , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Homeodomain Proteins/biosynthesis , Humans , Ligands , Male , Nanog Homeobox Protein , Neoplasms, Germ Cell and Embryonal/genetics , Neoplasms, Germ Cell and Embryonal/metabolism , Neoplasms, Germ Cell and Embryonal/pathology , Octamer Transcription Factor-3/biosynthesis , Positive Regulatory Domain I-Binding Factor 1 , Proto-Oncogene Proteins c-kit/biosynthesis , Repressor Proteins/biosynthesis , Seminoma/genetics , Seminoma/pathology , Signal Transduction , Testicular Neoplasms/genetics , Testicular Neoplasms/metabolism , Testicular Neoplasms/pathology
12.
Semin Cell Dev Biol ; 20(5): 607-19, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19447185

ABSTRACT

Adult fertility requires appropriate and coordinated instruction of somatic and germ cell activity during lineage specification, development and maturation. Driven by alterations in the complement of nuclear proteins such as transcription factors and chromatin remodelling components, these events proceed by sequential changes in gene expression in response to a myriad of signalling cues. Controlled access of proteins to the nucleus is a key driver of developmental switches. This review discusses key examples of regulated nucleocytoplasmic transport during mammalian gametogenesis and the mechanisms underpinning these transport events, focusing on examples critical for the establishment of fertility.


Subject(s)
Active Transport, Cell Nucleus , Gametogenesis , Mammals/metabolism , Animals , Humans , Nucleocytoplasmic Transport Proteins/metabolism
13.
Hum Reprod ; 23(2): 412-20, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18077314

ABSTRACT

BACKGROUND: The cellular sites of activin action and its regulation in the normal and dysfunctional adult human testis are unknown. METHODS: Activin type I (ALK2 and ALK4) and type II (ActRIIA and ActRIIB) receptors were detected using immunohistochemistry on Bouins fixed sections of normal, carcinoma in situ (CIS), seminoma, non-seminoma and gonadotropin-deprived human testis. ActRIIA mRNA was localized by in situ hybridization. RESULTS: ALK2, ALK4 and ActRIIB proteins were observed in Sertoli cells, spermatogonia and some spermatocytes within normal and gonadotropin-suppressed adult human testis; all three receptor subunits were also detected in CIS, seminoma and non-seminoma cells. ActRIIA immunoreactivity was faint to absent in the normal testis and in CIS and non-seminoma cells, whereas some seminoma cells displayed a strong signal. Also in contrast to the normal testis, a majority of spermatogonia and Sertoli cells in gonadotropin-deprived samples exhibited a strong ActRIIA immunohistochemical and in situ hybridization signal. CONCLUSIONS: Spermatogonia and Sertoli cells appear as the primary targets of activin action in the adult human testis. Changes in testicular function associated with altered hormone levels may enhance ActRIIA mRNA and protein synthesis, thus modifying signalling by activin or other TGFbeta ligands within specific cells of the seminiferous epithelium.


Subject(s)
Activin Receptors/metabolism , Carcinoma in Situ/metabolism , Seminoma/metabolism , Testicular Neoplasms/metabolism , Testis/metabolism , Activin Receptors/genetics , Adolescent , Adult , Drug Synergism , Gonadotropins/antagonists & inhibitors , Humans , Immunohistochemistry , In Situ Hybridization , Male , Medroxyprogesterone Acetate/pharmacology , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Sertoli Cells/metabolism , Spermatocytes/metabolism , Spermatogonia/metabolism , Testosterone/pharmacology , Tissue Distribution
14.
Int J Androl ; 30(4): 377-84; discussion 384, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17705810

ABSTRACT

Regulated transforming growth factor-beta (TGFbeta) superfamily signalling is an integral part of normal testicular development and the processes that enable the production of fertile sperm. Through shared utilization of receptors, signal transduction components and inhibitors, many ligands in this family exhibit functional overlaps; this facet of their function is critical to understand because these ligands are often co-expressed and, hence, they may compete with or compensate for one another, depending on the specific cellular context. This review describes particular germ cell maturation steps governed by bone morphogenetic proteins, glial cell line-derived neurotrophic factor and activins, focusing on data predominantly from rodent studies that implicate activin and other family members in modulation of gonocyte and spermatogonial stem cell development. We also review knowledge of the TGFbeta superfamily signalling components in the human testis, exploring their potential impact on the processes associated with disrupted gonocyte development and an enhanced risk of testicular cancer.


Subject(s)
Gonadal Dysgenesis/physiopathology , Spermatogenesis/physiology , Testicular Neoplasms/physiopathology , Transforming Growth Factor beta/physiology , Animals , Female , Male , Mice , Sertoli Cells/physiology , Sex Characteristics , Signal Transduction
15.
Int J Androl ; 30(4): 265-74; discussion 274, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17573852

ABSTRACT

Geneticists have a long history of studying reproduction in the fruitfly, Drosophila melanogaster, and in recent years it has become apparent that many of the genes that regulate invertebrate reproduction have been conserved through vertebrate evolution. As with other higher eukaryotes, spermatogenesis in Drosophila is characterized by a regenerative germline stem cell population that divides asymmetrically to produce mitotic spermatogonia which will eventually differentiate into spermatocytes. Germline tumours consisting of undifferentiated germ cells have been associated with both loss-of-function mutations and ectopic gene expression. While the genesis of these tumours may not be identical to human germ cell tumours many of the genes that regulate stem cell proliferation and aberrant over-proliferation in the Drosophila testis provide candidate molecules that may underlie the genetic programmes that contribute to human testicular oncogenesis.


Subject(s)
Drosophila melanogaster/physiology , Spermatogenesis/physiology , Testicular Neoplasms/physiopathology , Animals , Female , Humans , Male , Mitosis , Ovum/physiology , Reproduction , Spermatogonia/cytology , Testis/growth & development , Testis/physiology
16.
Reproduction ; 133(4): 753-61, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17504919

ABSTRACT

We present a detailed study of the expression pattern of WD repeat and SOCS box-containing 2 (Wsb2) in mouse embryonic and adult gonads. Wsb2 was previously identified in a differential screen aimed at identifying the genes involved in male- and female-specific gonadal development. Wsb2 expression was analysed during mouse gonadogenesis by real-time PCR, whole-mount and section in situ hybridisation and immunofluorescence. Wsb2 mRNA expression was initially detected in gonads of both sexes from 11.5 days post coitum (dpc) until 12.0 dpc. By 12.5 dpc and thereafter, Wsb2 expression rapidly decreased in the female, while persisting in the male gonads. In foetal, newborn and juvenile testes, Wsb2 mRNA and protein were readily detected in the seminiferous cords within both Sertoli and germ cells. Wsb2 mRNA was present in spermatogonia, spermatocytes and in Sertoli cells of the adult mouse testis. The differential expression of Wsb2 in male versus female embryonic gonads suggests some male-specific role in gonad development, and its expression in the first wave of spermatogenesis indicates a role in germ cells. Real-time analysis of adult mouse testis tubules cultured in the presence of the Hedgehog signalling inhibitor, cyclopamine, showed a downregulation of Wsb2 mRNA after treatment which suggests that Wsb2 may be a target of Hedgehog signalling.


Subject(s)
Carrier Proteins/genetics , RNA, Messenger/analysis , Suppressor of Cytokine Signaling Proteins/genetics , Testis/embryology , Testis/metabolism , Amino Acid Sequence , Animals , Base Sequence , Carrier Proteins/analysis , Chickens , Female , Gene Expression , Gene Expression Regulation, Developmental , Gestational Age , Humans , In Situ Hybridization/methods , Male , Mice , Molecular Sequence Data , Ovary/embryology , Ovary/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Sequence Alignment , Sertoli Cells/metabolism , Sex Differentiation/genetics , Spermatozoa/metabolism , Suppressor of Cytokine Signaling Proteins/analysis , Teratogens/pharmacology , Testis/drug effects , Tissue Culture Techniques , Veratrum Alkaloids/pharmacology
17.
Curr Genomics ; 8(5): 323-34, 2007 Aug.
Article in English | MEDLINE | ID: mdl-19384428

ABSTRACT

Access to nuclear genes in eukaryotes is provided by members of the importin (IMP) superfamily of proteins, which are of alpha- or beta-types, the best understood nuclear import pathway being mediated by a heterodimer of an IMP alpha and IMP beta1. IMP alpha recognises specific targeting signals on cargo proteins, while IMP beta1 mediates passage into, and release within, the nucleus by interacting with other components of the transport machinery, including the monomeric guanine nucleotide binding protein Ran. In this manner, hundreds of different proteins can be targeted specifically into the nucleus in a tightly regulated fashion. The IMP alpha gene family has expanded during evolution, with only a single IMP alpha (Srp1p) gene in budding yeast, and three (IMP alpha1, 2/pendulin and 3) and five (IMP alpha1, -2, -3, -4 and -6) IMP alpha genes in Drosophila melanogaster and mouse respectively, which fall into three phylogenetically distinct groups. The fact that IMP alpha3 and IMP alpha2 are only present in metazoans implies that they emerged during the evolution of multicellular animals to perform specialised roles in particular cells and tissues. This review describes what is known of the IMP alpha gene family in mouse and in D. melanogaster, including a comparitive examination of their mRNA expression profiles in a highly differentiated tissue, the testis. The clear implication of their highly regulated synthesis during the course of spermatogenesis is that the different IMP alphas have distinct expression patterns during cellular differentiation, implying tissue/cell type-specific roles.

18.
J Endocrinol ; 190(2): 331-40, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16899566

ABSTRACT

Production and regulation of activin A and inhibin B during the cycle of the seminiferous epithelium were investigated in adult rats. Immunohistochemistry localised the activin beta(A)-subunit to the Sertoli cell cytoplasm, with much weaker expression in spermatocytes and spermatids. Both activin A and inhibin B, measured by ELISA were secreted by, seminiferous tubule fragments over 72 h in culture. Activin A was secreted in a cyclic manner with peak secretion from tubules isolated at stage VIII. Tubules collected during stage VI produced the least activin A. Inhibin B secretion was highest from stage IX-I tubules and lowest from stage VII tubules. Addition of interleukin-1beta (IL-1beta) had relatively little effect on activin A or inhibin B secretion in culture. In contrast, the peak secretion of activin A by stage VIII tubules was blocked by co-incubation with an excess of human recombinant IL-1 receptor antagonist, whereas inhibin B secretion increased slightly. Dibutyryl cAMP stimulated activin A secretion by late stage VII and VIII tubules and stimulated inhibin B across all stages. These data indicate that activin A and inhibin B are cyclically regulated within the seminiferous epithelium, with endogenous IL-1 (presumably IL-1alpha produced by the Sertoli cells), responsible for a peak of activin A production subsequent to sperm release at stage VIII. These data provide direct evidence that production of activin A and inhibin B by the Sertoli cell is locally modulated by IL-1alpha , in addition to FSH/cAMP, under the influence of the developing spermatogenic cells.


Subject(s)
Activins/biosynthesis , Seminiferous Epithelium/metabolism , Spermatogenesis/physiology , Activins/analysis , Animals , Bucladesine/pharmacology , Cytoplasm/chemistry , Enzyme-Linked Immunosorbent Assay/methods , Immunohistochemistry/methods , Inhibins/analysis , Inhibins/biosynthesis , Interleukin 1 Receptor Antagonist Protein , Interleukin-1/metabolism , Male , Rats , Rats, Sprague-Dawley , Seminiferous Epithelium/chemistry , Sertoli Cells/chemistry , Sialoglycoproteins/pharmacology , Spermatozoa/chemistry , Stimulation, Chemical , Tissue Culture Techniques
19.
Dev Dyn ; 234(4): 1026-33, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16245330

ABSTRACT

In an effort to understand the mechanisms that underpin gonadal differentiation at the time of sex determination, we identified a cDNA encoding a putative novel testis expressed scavenger receptor, Tesr. Based on its domain structure, we hypothesize that the function of Tesr is similar to that of other scavenger receptors that play roles in phagocytosis of apoptotic cells, cell-cell adhesion, and defense. Tesr mRNA was detected in fetal mouse gonads of both sexes at 11.5 days post coitum (dpc). From 12.0 dpc, Tesr expression rapidly decreased in the female and was maintained in the male. Expression was seen in embryonic mouse sites other than the testis, such as in brain, eye, head, heart, neural arch, and cartilage primordium. Tesr expression in the newborn testis was faint to undetectable, but it increased from 2 days postpartum (dpp) until 15 dpp and was found in a subset of interstitial cells and in germ and Sertoli cells. Tesr mRNA in the adult mouse testis was observed in Sertoli cells, spermatogonia, spermatocytes, round spermatids, and in a subset of interstitial cells. We conclude that Tesr is differentially expressed in the male vs. female embryonic gonad and is expressed in both the ovary and the testes postnatally after 2 dpp.


Subject(s)
Gene Expression Regulation, Developmental , RNA, Messenger/metabolism , Receptors, Scavenger/genetics , Receptors, Scavenger/metabolism , Sex Differentiation/genetics , Testis/embryology , Testis/metabolism , Age Factors , Amino Acid Sequence , Animals , DNA Primers , Female , In Situ Hybridization , Male , Mice , Molecular Sequence Data , Ovary/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Sex Factors
20.
Ann N Y Acad Sci ; 1061: 173-82, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16467266

ABSTRACT

Spermatogenesis requires progression of germ line stem cells through a precisely ordered differentiation pathway to form spermatozoa. Diverse and dynamic signals from the transforming growth factor-beta (TGF-beta) superfamily influence many stages of germ cell development. For example, interactions between several TGF-beta superfamily ligands (bone morphogenetic proteins, activin, and glial-derived neurotrophic growth factor [GDNF]) appear to govern the onset of spermatogenesis, and we are exploring how germ cells interpret these competing signals. We examined the in vivo impact of activin on testis development using two mouse models, the inhba-/- mouse (which lacks the gene encoding the activin A subunit and dies at birth) and BK mice, with inhbb (encoding the activin betaB subunit) replacing inhba (which survive to adulthood and show delayed fertility onset in males). Distinct effects on Sertoli cell and germ cell populations during fetal and early postnatal development were measured. We recognize that specific proteins, including downstream targets of TGF-beta signals, such as Smads, must move into the nucleus to implement the gene transcription changes required for development. We hypothesized that changes at the level of cellular nuclear transport machinery may be required to mediate this. Examination of proteins involved in classical nuclear import, the importins, revealed that each importin has a developmentally regulated expression pattern in male germ cells. Because each importin binds a selected range of cargo proteins and mediates their nucleocytoplasmic passage, our findings suggest that each importin ferries cargo required for discrete stages of spermatogenesis.


Subject(s)
Spermatogenesis/physiology , Spermatozoa/growth & development , Testis/embryology , Testis/growth & development , Activins/metabolism , Activins/pharmacology , Animals , Biological Transport , Cell Differentiation , Cell Nucleus/metabolism , Gene Expression Regulation, Developmental , Karyopherins/metabolism , Male , Mice , Models, Biological , Signal Transduction , Spermatozoa/physiology , Testis/cytology , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...