Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
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(5): 703-711, 2019 09.
Article in English | MEDLINE | ID: mdl-31322318

ABSTRACT

BACKGROUND: The interface between the epididymis and the immune system is implicated in many male reproductive pathologies. The resident immune cell populations and immune-environment within the epididymis are significantly different from the testis, which is an immune-privileged site. Moreover, the immune cell subsets and immunological responses between different regions of the epididymis vary considerably. The cauda epididymis is more susceptible to autoimmune responses than the caput in rodent models of active immunization or suppressed immune tolerance, and in men with congenital or physical damage to the reproductive tract. Activins are members of the transforming growth factor-ß family of cytokines that are crucial for testis and epididymal development; however, they also have complex immunoregulatory properties and may play an essential role in the regulation of immunity in the reproductive tract. MATERIALS AND METHODS: Our recent research and relevant publications by other researchers identified following a PubMed search are reviewed. RESULTS: The caput epididymis displays elevated endogenous expression of activins A and B and the immunoregulatory gene, indoleamine-2,3-dioxygenase, co-existing with an extensive population of intra-epithelial and interstitial macrophages and dendritic cells, which appear to be involved in regulating tolerance against sperm antigens. The caput is also relatively resistant to inflammatory damage caused by autoimmunity or bacterial infection, but the cauda, which exhibits low activin expression and high levels of the activin-binding protein, follistatin, is highly susceptible to inflammatory damage. Paradoxically, inflammation in the cauda induces increased activin production, and inhibition of activin activity reduces inflammatory responses. Studies using mouse models with altered levels of activins and follistatin indicate a relationship between the activins and genes involved in inflammation and immunoregulation. CONCLUSION: The existing data indicate that activins play a complex role in controlling inflammation and immunity in the epididymis and vas deferens.


Subject(s)
Activins/metabolism , Epididymis/immunology , Epididymitis/pathology , Follistatin/metabolism , Vas Deferens/pathology , Animals , Epididymis/pathology , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Inhibin-beta Subunits/genetics , Inhibins/genetics , Male , Mice , Models, Animal , Vas Deferens/immunology
3.
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
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.
Reprod Fertil Dev ; 27(7): 985-99, 2015 Sep.
Article in English | MEDLINE | ID: mdl-24630125

ABSTRACT

Female mice lacking the follistatin gene but expressing a human follistatin-315 transgene (tghFST315) have reproductive abnormalities (reduced follicles, no corpora lutea and ovarian-uterine inflammation). We hypothesised that the absence of follistatin-288 causes the abnormal reproductive tract via both developmental abnormalities and abnormal ovarian activity. We characterised the morphology of oviducts and uteri in wild type (WT), tghFST315 and follistatin-knockout mice expressing human follistatin-288 (tghFST288). The oviducts and uteri were examined in postnatal Day-0 and adult mice (WT and tghFST315 only) using histology and immunohistochemistry. Adult WT and tghFST315 mice were ovariectomised and treated with vehicle, oestradiol-17ß (100ng injection, dissection 24h later) or progesterone (1mg×three daily injections, dissection 24h later). No differences were observed in the oviducts or uteri at birth, but abnormalities developed by adulthood. Oviducts of tghFST315 mice failed to coil, the myometrium was disorganised, endometrial gland number was reduced and oviducts and uteri contained abundant leukocytes. After ovariectomy, tghFST315 mice had altered uterine cell proliferation, and inflammation was maintained and exacerbated by oestrogen. These studies show that follistatin is crucial to postnatal oviductal-uterine development and function. Further studies differentiating the role of ovarian versus oviductal-uterine follistatin in reproductive tract function at different developmental stages are warranted.


Subject(s)
Follistatin/genetics , Oviducts/growth & development , Uterus/growth & development , Animals , Cell Proliferation/genetics , Endometrium/growth & development , Endometrium/metabolism , Estrogens/pharmacology , Female , Follistatin/metabolism , Gene Expression Regulation, Developmental , Mice , Mice, Knockout , Mice, Transgenic , Myometrium/growth & development , Myometrium/metabolism , Ovariectomy , Oviducts/diagnostic imaging , Oviducts/metabolism , Uterus/drug effects , Uterus/metabolism
7.
Anim Reprod Sci ; 151(3-4): 148-56, 2014 Dec 30.
Article in English | MEDLINE | ID: mdl-25458319

ABSTRACT

The onset of puberty depends on the attainment of critical body mass, so should also be affected by increases in the rate of accumulation of muscle and adipose tissue. Adipose tissue and reproduction are linked by leptin. For muscle, a link has not yet been identified, although one possibility is follistatin. We assessed the relationships among circulating concentrations of follistatin and leptin and the rates of growth and accumulation of muscle and fat during pubertal development in female sheep. We used 326 animals with known phenotypic values for live weight (LW), depths of eye muscle (EMD) and fat (FAT), and known breeding values at post-weaning age for body mass (PWT) and depths of eye muscle (PEMD) and fat (PFAT). Leptin concentration was positively correlated with values for EMD, PEMD, FAT, PFAT, LW and PWT (P<0.001), whereas follistatin concentration was negatively correlated with values for EMD and PWT (P<0.001), and PEMD (P<0.01) and FAT (P<0.05). Leptin concentration was negatively related to age and positively related to live weight at first oestrus and the proportion of females that attained puberty (P≤0.05), and to fertility and reproductive rate (P<0.01). Follistatin concentration was negatively related to live weight at first oestrus and to fertility (P<0.01) and reproductive rate (P<0.05). There were positive correlations (P<0.001) between muscle accumulation and leptin concentration, and between muscle accumulation and reproductive performance. We conclude that leptin and follistatin are probably both involved in effects of accelerated accumulation of muscle and adipose tissues on the onset of puberty.


Subject(s)
Body Composition , Fertility , Follistatin/blood , Leptin/blood , Sexual Maturation , Sheep, Domestic , Adipose Tissue/growth & development , Animals , Body Weight , Female , Muscle, Skeletal/growth & development , Sheep, Domestic/growth & development , Sheep, Domestic/metabolism
8.
Cytokine Growth Factor Rev ; 24(3): 285-95, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23541927

ABSTRACT

The activins, as members of the transforming growth factor-ß superfamily, are pleiotrophic regulators of cell development and function, including cells of the myeloid and lymphoid lineages. Clinical and animal studies have shown that activin levels increase in both acute and chronic inflammation, and are frequently indicators of disease severity. Moreover, inhibition of activin action can reduce inflammation, damage, fibrosis and morbidity/mortality in various disease models. Consequently, activin A and, more recently, activin B are emerging as important diagnostic tools and therapeutic targets in inflammatory and fibrotic diseases. Activin antagonists such as follistatin, an endogenous activin-binding protein, offer considerable promise as therapies in conditions as diverse as sepsis, liver fibrosis, acute lung injury, asthma, wound healing and ischaemia-reperfusion injury.


Subject(s)
Activins/physiology , Follistatin/metabolism , Activins/biosynthesis , Animals , Asthma/drug therapy , Disease Models, Animal , Female , Fibrosis , Follistatin/therapeutic use , Humans , Inflammation/drug therapy , Inhibins/physiology , Male , Wound Healing/drug effects
9.
Reprod Biomed Online ; 24(3): 364-74, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22285243

ABSTRACT

Follistatin, an inhibitor of activin A, has key regulatory roles in the female reproductive tract. Follistatin has two splice variants: FST288, largely associated with cell surfaces, and FST315, the predominant circulating form. The mechanism regulating uterine expression of these variants is unknown. Quantitative RT-PCR was used to measure expression of follistatin splice variants (Fst288, Fst315), the activin bA subunit (Inhba) and the inhibin a subunit (Inha) in uterine tissues during early pregnancy (days 1­4, preimplantation) and in response to exogenous 17b-oestradiol (single s.c. injection) and progesterone (three daily s.c. injections) in ovariectomized mice. Uterine Fst288, Fst315 and Inhba expression increased during early pregnancy, with greater increases in Fst315 relative to Fst288 suggesting differential regulation of these variants. Fst288, Fst315, Inhba and Inha all increased in response to progesterone treatment. Fst288, but not Fst315, mRNA decreased in response to 17b-oestradiol treatment, whereas Inhba increased. A comparison of the absolute concentrations of uterine follistatin mRNA using crossing thresholds indicated that both variants were more highly expressed in early pregnancy in contrast to the hormone treatment models. It is concluded that progesterone regulates uterine expression of both follistatin variants, as well as activin A, during early pregnancy in the mouse uterus


Subject(s)
Follistatin/metabolism , Gene Expression Regulation/drug effects , Progesterone/pharmacology , Uterus/drug effects , Animals , Estradiol/pharmacology , Female , Follistatin/chemistry , Follistatin/genetics , Inhibin-beta Subunits/genetics , Inhibin-beta Subunits/metabolism , Inhibins/genetics , Inhibins/metabolism , Mice , Pregnancy , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Uterus/metabolism
10.
Mol Hum Reprod ; 15(11): 757-61, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19602508

ABSTRACT

Identifying suitable housekeeping genes for quantitative RT-PCR in the uterus is problematic, as this tissue undergoes significant structural and functional alterations during the oestrous cycle and pregnancy in response to circulating hormones. The suitability of 18S rRNA as a housekeeping gene in mouse uterus was investigated by introducing an 'RNA spike' standard into the reverse transcription reaction. 18S rRNA levels increased by Day 4 of pregnancy and after progesterone administration in ovariectomized mice. We conclude that 18S rRNA is not a suitable housekeeping gene for quantitative RT-PCR analysis in progesterone-responsive tissues, and the RNA spiking method provides a suitable alternative.


Subject(s)
Progesterone/metabolism , RNA, Ribosomal, 18S/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Uterus/metabolism , Animals , Estradiol/pharmacology , Female , Mice , Ovariectomy , Pregnancy , Progesterone/pharmacology , Uterus/drug effects
11.
Mol Cell Endocrinol ; 307(1-2): 169-75, 2009 Aug 13.
Article in English | MEDLINE | ID: mdl-19524137

ABSTRACT

Bacterial lipopolysaccharide increased the production of interleukin 1alpha and activin A, and reduced production of inhibin B, in Sertoli cells from immature male rats measured by enzyme-linked immunosorbent assay (ELISA). The majority of immunoreactive interleukin 1alpha remained within the Sertoli cell, while both activin A and inhibin B were secreted. Lipopolysaccharide-stimulated expression of two interleukin 1alpha mRNA transcripts, measured by quantitative RT-PCR, but the levels of bioactive interleukin 1alpha in Sertoli cell extracts and medium, measured by in vitro bioassay, were comparatively low to undetectable. A specific antagonist of interleukin 1alpha had no effect on lipopolysaccharide-stimulated activin A or inhibin B responses. These data indicate that, in contrast to Sertoli cells from adult rats, lipopolysaccharide-induced regulation of activin A and inhibin B by prepubertal Sertoli cells does not involve secreted interleukin 1alpha. The data highlight the possibility of a role for intracellular interleukin 1alpha in the Sertoli cell response to inflammation, particularly in the immature testis.


Subject(s)
Activins/metabolism , Inhibins/metabolism , Interleukin-1alpha/metabolism , Lipopolysaccharides/pharmacology , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Activins/genetics , Animals , Biological Assay , Cell Extracts , Culture Media , Gene Expression Regulation/drug effects , Inhibins/genetics , Interleukin-1alpha/genetics , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats
12.
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
13.
J Endocrinol ; 187(1): 125-34, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16214948

ABSTRACT

The regulation of Sertoli cell activin A and inhibin B secretion during inflammation was investigated in vitro. Adult rat Sertoli cells were incubated with the inflammatory mediators, lipopolysaccharide (LPS), interleukin-1beta (IL-1beta), IL-6 and the IL-1 receptor antagonist (IL-1ra) over 48 h in culture. Activin A, inhibin B and IL-1alpha were measured in the culture medium by specific two-site ELISAs. Both IL-1beta- and LPS-stimulated activin A and inhibited inhibin B secretion. LPS also stimulated the production of IL-1alpha in the cultures. In contrast to IL-1beta, IL-6 had no effect on activin A, although it did have a significant inhibitory effect on inhibin B secretion. Ovine follicle-stimulating hormone (FSH) and the cAMP analogue dibutyryl cAMP opposed the actions of IL-1 and LPS by suppressing activin A and IL-1alpha secretion and by stimulating inhibin B. Blocking IL-1 activity in the cultures by addition of an excess of IL-1ra completely prevented the response of activin A to exogenous IL-1beta, and reduced the response to LPS by 50%. In the presence of IL-1ra, basal secretion of inhibin B was increased, but IL-1ra was unable to reverse the suppression of inhibin B by LPS. These data indicate the importance of both IL-1 isoforms in regulating secretion of activin A and inhibin B by mature Sertoli cells during inflammation. The data also establish that inflammation exerts its effects on activin A and inhibin B secretion via other pathways in addition to those mediated by IL-1, and that hormonal stimulation by FSH and cAMP moderates the Sertoli cell response to inflammation. Interference with the complex interactions between these cytokines and hormones may contribute to the disruption of reproductive function that can accompany infection and illness in men.


Subject(s)
Activins/metabolism , Inflammation Mediators/pharmacology , Inhibin-beta Subunits/metabolism , Inhibins/metabolism , Sertoli Cells/metabolism , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay/methods , Interleukin 1 Receptor Antagonist Protein , Interleukin-1/pharmacology , Interleukin-6/pharmacology , Lipopolysaccharides/pharmacology , Male , Rats , Rats, Sprague-Dawley , Sertoli Cells/drug effects , Sertoli Cells/immunology , Sialoglycoproteins/pharmacology , Stimulation, Chemical
14.
J Endocrinol ; 185(1): 99-110, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15817831

ABSTRACT

In several biological systems, the inhibin beta(A) homodimer activin A is stimulated by, and in turn, inhibits the action of interleukin (IL)-1 (both IL-1alpha and IL-1beta) and IL-6. The possibility that a similar regulatory relationship operates within the testis was investigated. Sertoli cells from immature (20-day-old) rats were cultured with human IL-1alpha or IL-1beta, human IL-6 and/or ovine FSH or dibutyryl cAMP. Activin A and the inhibin dimers, inhibin A and inhibin B, were measured by specific ELISA. Immunoreactive inhibin (ir-inhibin) was measured by RIA. Activin/inhibin subunit mRNA expression was measured by quantitative real-time PCR. Both IL-1 isoforms, but not IL-6, stimulated activin A secretion through increased synthesis of beta(A)-subunit mRNA. IL-1 also stimulated activin A secretion by testicular peritubular cells. In contrast to the effect on activin A, IL-1 suppressed inhibin beta(B)-subunit and, to a lesser extent, alpha-subunit mRNA expression, thereby reducing basal and FSH-stimulated inhibin B secretion by the Sertoli cells. Conversely, FSH inhibited basal activin A secretion and antagonised the stimulatory effects of IL-1. Dibutyryl cAMP partially inhibited the action of IL-1 on activin A secretion, but had no significant effect on basal activin A secretion. Secretion of inhibin A was low in all treatment groups. These data demonstrate that IL-1 and FSH/cAMP exert a reciprocal regulation of activin A and inhibin B synthesis and release by the Sertoli cell, and suggest a role for activin A as a potential feedback regulator of IL-1 and IL-6 activity in the testis during normal spermatogenesis and in inflammation.


Subject(s)
Activins/pharmacology , Follicle Stimulating Hormone/pharmacology , Inhibin-beta Subunits/pharmacology , Inhibins/pharmacology , Interleukin-1/pharmacology , Sertoli Cells/metabolism , Animals , Bucladesine/pharmacology , Cell Culture Techniques , Enzyme-Linked Immunosorbent Assay/methods , Feedback, Physiological , Interleukin-6/pharmacology , Male , RNA, Messenger/analysis , Rats , Reverse Transcriptase Polymerase Chain Reaction , Sertoli Cells/drug effects , Stimulation, Chemical
15.
Mol Cell Endocrinol ; 225(1-2): 57-64, 2004 Oct 15.
Article in English | MEDLINE | ID: mdl-15451568

ABSTRACT

The role of the inhibins, activins and follistatins in testicular function are being more clearly defined following studies describing the cellular localisation of these proteins to the testis and the availability of specific assay systems enabling measurement of these proteins. Taken together with the results of targetted gene inactivation experiments, several concepts emerge. Inhibin B is predominantly produced by the Sertoli cell in many adult male mammals whereas there is a perinatal peak of inhibin A in the rat. In contrast, activin A has its highest concentrations in the immediate post-natal period during which it is involved in the developmental regulation of both germ cells and Sertoli cells being modulated by follistatin. Activin A levels are considerably lower in the adult testis but Sertoli cell production is stimulated by interleukin-1 and inhibited by FSH. Little is known about the production of activin B due to the absence of a suitable assay but the beta(B) subunit mRNA is expressed in germ cells and Sertoli cells and is stage-dependent. This pattern of expression suggest that it may be involved in autocrine or paracrine actions within the seminiferous epithelium.


Subject(s)
Gonadal Hormones/physiology , Testis/physiology , Activins/genetics , Activins/metabolism , Activins/physiology , Animals , Follistatin/genetics , Follistatin/metabolism , Follistatin/physiology , Gene Expression Regulation/physiology , Gonadal Hormones/genetics , Humans , Inhibins/physiology , Male
16.
Hum Reprod Update ; 8(6): 529-41, 2002.
Article in English | MEDLINE | ID: mdl-12498423

ABSTRACT

The regulation of reproductive processes involves a complex network of communication systems between the brain, endocrine organs, the gonads and other reproductive tissues. Classically, our understanding has focused on the role of endocrine hormones, but more recently interest has also dwelt on the paracrine and autocrine regulation of these cell systems. In this review, the structure and physiology of the inhibins, activins and follistatin are discussed in terms of the evidence supporting their role as endocrine hormones, and how they might function as paracrine factors within the pituitary, gonad and associated tissues. With the advent of more specific techniques and assays for their measurement, the potential of inhibins, activins and follistatin as clinical markers of reproductive function and in the screening of various pathologies is also evaluated.


Subject(s)
Activins/physiology , Follistatin/physiology , Inhibins/physiology , Reproduction , Animals , Female , Follicle Stimulating Hormone/metabolism , Humans , Male , Ovary , Pregnancy , Testis
17.
Mol Hum Reprod ; 8(6): 518-24, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12029068

ABSTRACT

Macrophages are numerous in the testicular interstitial tissue under normal conditions and increase during inflammation. The mechanisms involved are poorly characterized. Expression of the macrophage-regulating cytokines monocyte chemoattractant protein (MCP)-1 and macrophage colony-stimulating factor (M-CSF) was examined in the adult rat testis before and after an i.p. injection of an inflammatory stimulus, lipopolysaccharide (LPS). In the normal testis, M-CSF was readily observed using Northern blot and Western blot analysis. In contrast, MCP-1 was not detectable by Northern blot in the normal testis, but was detected using RT-PCR amplification and a sensitive ELISA. After LPS treatment, testicular MCP-1 mRNA and protein expression increased dramatically (up to 400-fold). In-situ hybridization for MCP-1 revealed that production was confined to the interstitium of the inflamed testis, in Leydig cells, peritubular cells, perivascular cells and monocyte-like macrophages, but not in tissue-resident macrophages. Unlike MCP-1, M-CSF mRNA and protein expression in the testis increased only marginally, if at all, after LPS treatment. These results suggest that MCP-1 stimulates the increase in intratesticular macrophages that accompanies LPS-induced inflammation in vivo. Together with M-CSF, MCP-1 may also play a role in maintaining the resident macrophage population of the normal testis.


Subject(s)
Chemokine CCL2/metabolism , Inflammation/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Testis/physiology , Animals , Chemokine CCL2/genetics , In Situ Hybridization , Inflammation/chemically induced , Inflammation/genetics , Lipopolysaccharides/pharmacology , Liver/physiology , Macrophage Colony-Stimulating Factor/genetics , Macrophages/cytology , Macrophages/metabolism , Male , Monocytes/cytology , Monocytes/metabolism , Rats , Rats, Sprague-Dawley , Testis/cytology , Testis/drug effects , Testis/pathology
18.
Mol Cell Endocrinol ; 180(1-2): 155-62, 2001 Jun 30.
Article in English | MEDLINE | ID: mdl-11451586

ABSTRACT

The inflammatory cascade is a multifactorial process regulated by interwoven cytokine and growth factor networks. This review summarizes the emerging evidence that implicate activin A and follistatin in inflammatory processes. Our recent studies have determined that activin A is released early in the cascade of circulatory cytokines during systemic inflammatory episodes, roughly coincident with tumour necrosis factor (TNF)-alpha and before interleukin (IL)-6 and follistatin. The source(s) of this activin A are not yet established, but prime candidates are monocytes/macrophages, other immune cell types or vascular endothelial cells. Clinical data are limited, but activin beta(A) subunit mRNA or activin A protein is elevated in inflammatory bowel diseases and inflammatory arthropathies, and circulating concentrations of follistatin are elevated in patients with sepsis. In more mechanistic approaches, in vitro studies show that activin A can have both pro- and anti-inflammatory actions on key inflammatory mediators such as TNFalpha, IL-1beta and IL-6. Furthermore, there is emerging understanding of how the intracellular signaling pathway for activin A, incorporating Smads, may interact with and be modulated by other key regulatory cytokines and growth factors.


Subject(s)
Activins/pharmacology , Activins/physiology , Inflammation/metabolism , Inhibin-beta Subunits/physiology , Activins/metabolism , Animals , Drug Interactions , Follistatin , Humans , Inhibin-beta Subunits/metabolism , Inhibin-beta Subunits/pharmacology , Signal Transduction/drug effects
19.
J Endocrinol ; 168(1): 193-201, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11139783

ABSTRACT

A single intraperitoneal injection of lipopolysaccharide (LPS) causes a biphasic suppression of testicular steroidogenesis in adult rats, with inhibition at 6 h and 18-24 h after injection. The inhibition of steroidogenesis is independent of the reduction in circulating LH that also occurs after LPS treatment, indicating a direct effect of inflammation at the Leydig cell level. The relative contributions to this inhibition by intratesticular versus systemic responses to inflammation, including the adrenal glucocorticoids, was investigated in this study. Adult male Wistar rats (eight/group) received injections of LPS (0.1 mg/kg i.p.), dexamethasone (DEX; 50 microg/kg i.p.), LPS and DEX, or saline only (controls), and were killed 6 h, 18 h and 72 h later. Treatment with LPS stimulated body temperature and serum corticosterone levels measured 6 h later. Administration of DEX had no effect on body temperature, but suppressed serum corticosterone levels. At the dose used in this study, DEX alone had no effect on serum LH or testosterone at any time-point. Expression of mRNA for interleukin-1beta (IL-1beta), the principal inflammatory cytokine, was increased in both testis and liver of LPS-treated rats. Serum LH and testosterone levels were considerably reduced at 6 h and 18 h after LPS treatment, and had not completely recovered by 72 h. At 6 h after injection, DEX inhibited basal IL-1beta expression and the LPS-induced increase of IL-1beta mRNA levels in the liver, but had no effect on IL-1beta in the testis. The effects of DEX on IL-1beta levels in the liver were no longer evident by 18 h. In LPS-treated rats, DEX caused a significant reversal of the inhibition of serum LH and testosterone at 18 h, although not at 6 h or 72 h. Accordingly, DEX inhibited the systemic inflammatory response, but had no direct effect on either testicular steroidogenesis or intra-testicular inflammation, at the dose employed. These data suggest that the inhibition of Leydig cell steroidogenesis at 6 h after LPS injection, which was not prevented by co-administration of DEX, is most likely due to direct actions of LPS at the testicular level. In contrast, the later Leydig cell inhibition (at 18 h) may be attributable to extra-testicular effects of LPS, such as increased circulating inflammatory mediators or the release of endogenous glucocorticoids, that were inhibited by DEX treatment. These data indicate that the early and late phases of Leydig cell inhibition following LPS administration are due to separate mechanisms.


Subject(s)
Dexamethasone/therapeutic use , Glucocorticoids/therapeutic use , Leydig Cells/metabolism , Orchitis/drug therapy , Testosterone/metabolism , Analysis of Variance , Animals , Blotting, Northern/methods , Corticosterone/blood , Interleukin-1/genetics , Leydig Cells/drug effects , Leydig Cells/immunology , Lipopolysaccharides , Liver/immunology , Luteinizing Hormone/blood , Male , Orchitis/blood , Orchitis/immunology , RNA, Messenger/analysis , Rats , Rats, Wistar , Testosterone/blood , Time Factors
20.
Biol Reprod ; 63(5): 1285-93, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11058531

ABSTRACT

In vitro data have indicated that nitric oxide (NO) inhibits Leydig cell testosterone production, suggesting that NO may play a role in the suppression of steroidogenesis and spermatogenic function during inflammation. Consequently, we investigated expression of the inflammation-inducible isoform of NO synthase (iNOS) in the inflamed adult rat testis and the ability of a broad-spectrum inhibitor of NO production, L-nitro-L-arginine methyl ester, to prevent Leydig cell dysfunction during inflammation. Unexpectedly, immunohistochemical and mRNA data established that iNOS is expressed constitutively in Leydig cells and in a stage-specific manner in Sertoli, peritubular, and spermatogenic cells in the normal testis. Expression was increased in a dose-dependent manner in all these cell types during lipopolysaccharide (LPS)-induced inflammation. In noninflamed testes, treatment with the NO synthase inhibitor reduced testicular interstitial fluid formation and testosterone production without any effect on serum LH levels. Administration of the inhibitor did not prevent the suppression of testicular interstitial fluid and testosterone production that occurs within 6 h after LPS treatment. Collectively, these data indicate a novel role for iNOS in autocrine or paracrine regulation of the testicular vasculature, Leydig cell steroidogenesis, and spermatogenesis in the normal testis. The data suggest that increased NO is not the major cause of acute Leydig cell dysfunction in the LPS-treated inflammation model, although a role for NO in this process cannot be excluded, particularly at other time points. Moreover, up-regulation of iNOS may contribute to the seminiferous epithelium damage caused by LPS-induced inflammation.


Subject(s)
Infertility, Male/enzymology , Inflammation/enzymology , Nitric Oxide Synthase/physiology , Testis/enzymology , Animals , Blotting, Northern , Databases, Factual , Hormones/blood , Immunohistochemistry , Indicators and Reagents , Lipopolysaccharides/pharmacology , Male , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Rats , Reverse Transcriptase Polymerase Chain Reaction , Testis/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...