1
|
Amarilla MS, Glienke L, Munduruca Pires T, Sobarzo CM, Oxilia HG, Fulco MF, Rodríguez Peña M, Maio MB, Ferrer Viñals D, Lustig L, Jacobo PV, Theas MS. Impaired Spermatogenesis in Infertile Patients with Orchitis and Experimental Autoimmune Orchitis in Rats. BIOLOGY 2024; 13:278. [PMID: 38666890 PMCID: PMC11048156 DOI: 10.3390/biology13040278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/10/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024]
Abstract
Experimental autoimmune orchitis (EAO) is a well-established rodent model of organ-specific autoimmunity associated with infertility in which the testis immunohistopathology has been extensively studied. In contrast, analysis of testis biopsies from infertile patients associated with inflammation has been more limited. In this work, testicular biopsies from patients with idiopathic non-obstructive azoospermia diagnosed with hypospermatogenesis (HypoSp) [mild: n = 9, and severe: n = 11], with obstructive azoospermia and complete Sp (spermatogenesis) (control group, C, n = 9), and from Sertoli cell-only syndrome (SCOS, n = 9) were analyzed for the presence of immune cells, spermatogonia and Sertoli cell (SCs) alterations, and reproductive hormones levels. These parameters were compared with those obtained in rats with EAO. The presence of increased CD45+ cells in the seminiferous tubules (STs) wall and lumen in severe HypoSp is associated with increased numbers of apoptotic meiotic germ cells and decreased populations of undifferentiated and differentiated spermatogonia. The SCs showed an immature profile with the highest expression of AMH in patients with SCOS and severe HypoSp. In SCOS patients, the amount of SCs/ST and Ki67+ SCs/ST increased and correlated with high serum FSH levels and CD45+ cells. In the severe phase of EAO, immune cell infiltration and apoptosis of meiotic germ cells increased and the number of undifferentiated and differentiated spermatogonia was lowest, as previously reported. Here, we found that orchitis leads to reduced sperm number, viability, and motility. SCs were mature (AMH-) but increased in number, with Ki67+ observed in severely damaged STs and associated with the highest levels of FSH and inflammatory cells. Our findings demonstrate that in a scenario where a chronic inflammatory process is underway, FSH levels, immune cell infiltration, and immature phenotypes of SCs are associated with severe changes in spermatogenesis, leading to azoospermia. Furthermore, AMH and Ki67 expression in SCs is a distinctive marker of severe alterations of STs in human orchitis.
Collapse
Affiliation(s)
- María Sofía Amarilla
- Instituto de Investigaciones Biomédicas (INBIOMED), CONICET-Universidad de Buenos Aires, Paraguay 2155, Piso 10, Laboratorio 10, Ciudad Autónoma de Buenos Aires C1421ABG, Argentina; (L.G.); (T.M.P.); (C.M.S.); (M.B.M.); (D.F.V.); (L.L.)
| | - Leilane Glienke
- Instituto de Investigaciones Biomédicas (INBIOMED), CONICET-Universidad de Buenos Aires, Paraguay 2155, Piso 10, Laboratorio 10, Ciudad Autónoma de Buenos Aires C1421ABG, Argentina; (L.G.); (T.M.P.); (C.M.S.); (M.B.M.); (D.F.V.); (L.L.)
- Departamento de Biología Celular, Facultad de Medicina, Universidad de Buenos Aires, Cátedra II de Histología, Paraguay 2155, Ciudad Autónoma de Buenos Aires C1421ABG, Argentina;
| | - Thaisy Munduruca Pires
- Instituto de Investigaciones Biomédicas (INBIOMED), CONICET-Universidad de Buenos Aires, Paraguay 2155, Piso 10, Laboratorio 10, Ciudad Autónoma de Buenos Aires C1421ABG, Argentina; (L.G.); (T.M.P.); (C.M.S.); (M.B.M.); (D.F.V.); (L.L.)
- Departamento de Biología Celular, Facultad de Medicina, Universidad de Buenos Aires, Cátedra II de Histología, Paraguay 2155, Ciudad Autónoma de Buenos Aires C1421ABG, Argentina;
| | - Cristian Marcelo Sobarzo
- Instituto de Investigaciones Biomédicas (INBIOMED), CONICET-Universidad de Buenos Aires, Paraguay 2155, Piso 10, Laboratorio 10, Ciudad Autónoma de Buenos Aires C1421ABG, Argentina; (L.G.); (T.M.P.); (C.M.S.); (M.B.M.); (D.F.V.); (L.L.)
- Departamento de Biología Celular, Facultad de Medicina, Universidad de Buenos Aires, Cátedra II de Histología, Paraguay 2155, Ciudad Autónoma de Buenos Aires C1421ABG, Argentina;
| | - Hernán Gustavo Oxilia
- Departamento de Biología Celular, Facultad de Medicina, Universidad de Buenos Aires, Cátedra II de Histología, Paraguay 2155, Ciudad Autónoma de Buenos Aires C1421ABG, Argentina;
- Anatomía Patológica, Hospital General de Agudos Parmenio Piñero, Varela 1301, Ciudad Autónoma de Buenos Aires C1406ELA, Argentina
| | - María Florencia Fulco
- Hospital de Clínicas General San Martín, Av. Córdoba 2351 (C1120AAR), Ciudad Autónoma de Buenos Aires C1421ABG, Argentina; (M.F.F.); (M.R.P.)
| | - Marcelo Rodríguez Peña
- Hospital de Clínicas General San Martín, Av. Córdoba 2351 (C1120AAR), Ciudad Autónoma de Buenos Aires C1421ABG, Argentina; (M.F.F.); (M.R.P.)
| | - María Belén Maio
- Instituto de Investigaciones Biomédicas (INBIOMED), CONICET-Universidad de Buenos Aires, Paraguay 2155, Piso 10, Laboratorio 10, Ciudad Autónoma de Buenos Aires C1421ABG, Argentina; (L.G.); (T.M.P.); (C.M.S.); (M.B.M.); (D.F.V.); (L.L.)
| | - Denisse Ferrer Viñals
- Instituto de Investigaciones Biomédicas (INBIOMED), CONICET-Universidad de Buenos Aires, Paraguay 2155, Piso 10, Laboratorio 10, Ciudad Autónoma de Buenos Aires C1421ABG, Argentina; (L.G.); (T.M.P.); (C.M.S.); (M.B.M.); (D.F.V.); (L.L.)
| | - Livia Lustig
- Instituto de Investigaciones Biomédicas (INBIOMED), CONICET-Universidad de Buenos Aires, Paraguay 2155, Piso 10, Laboratorio 10, Ciudad Autónoma de Buenos Aires C1421ABG, Argentina; (L.G.); (T.M.P.); (C.M.S.); (M.B.M.); (D.F.V.); (L.L.)
- Departamento de Biología Celular, Facultad de Medicina, Universidad de Buenos Aires, Cátedra II de Histología, Paraguay 2155, Ciudad Autónoma de Buenos Aires C1421ABG, Argentina;
| | - Patricia Verónica Jacobo
- Instituto de Investigaciones Biomédicas (INBIOMED), CONICET-Universidad de Buenos Aires, Paraguay 2155, Piso 10, Laboratorio 10, Ciudad Autónoma de Buenos Aires C1421ABG, Argentina; (L.G.); (T.M.P.); (C.M.S.); (M.B.M.); (D.F.V.); (L.L.)
- Departamento de Biología Celular, Facultad de Medicina, Universidad de Buenos Aires, Cátedra II de Histología, Paraguay 2155, Ciudad Autónoma de Buenos Aires C1421ABG, Argentina;
| | - María Susana Theas
- Instituto de Investigaciones Biomédicas (INBIOMED), CONICET-Universidad de Buenos Aires, Paraguay 2155, Piso 10, Laboratorio 10, Ciudad Autónoma de Buenos Aires C1421ABG, Argentina; (L.G.); (T.M.P.); (C.M.S.); (M.B.M.); (D.F.V.); (L.L.)
- Departamento de Biología Celular, Facultad de Medicina, Universidad de Buenos Aires, Cátedra II de Histología, Paraguay 2155, Ciudad Autónoma de Buenos Aires C1421ABG, Argentina;
| |
Collapse
|
2
|
Kasimanickam VR, Kasimanickam RK. In Silico Analysis of miRNA-Mediated Genes in the Regulation of Dog Testes Development from Immature to Adult Form. Animals (Basel) 2023; 13:ani13091520. [PMID: 37174557 PMCID: PMC10177090 DOI: 10.3390/ani13091520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 04/22/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
High-throughput in-silico techniques help us understand the role of individual proteins, protein-protein interaction, and their biological functions by corroborating experimental data as epitomized biological networks. The objective of this investigation was to elucidate the association of miRNA-mediated genes in the regulation of dog testes development from immature to adult form by in-silico analysis. Differentially expressed (DE) canine testis miRNAs between healthy immature (2.2 ± 0.13 months; n = 4) and mature (11 ± 1.0 months; n = 4) dogs were utilized in this investigation. In silico analysis was performed using miRNet, STRING, and ClueGo programs. The determination of mRNA and protein expressions of predicted pivotal genes and their association with miRNA were studied. The results showed protein-protein interaction for the upregulated miRNAs, which revealed 978 enriched biological processes GO terms and 127 KEGG enrichment pathways, and for the down-regulated miRNAs revealed 405 significantly enriched biological processes GO terms and 72 significant KEGG enrichment pathways (False Recovery Rate, p < 0.05). The in-silico analysis of DE-miRNA's associated genes revealed their involvement in the governing of several key biological functions (cell cycle, cell proliferation, growth, maturation, survival, and apoptosis) in the testis as they evolve from immature to adult forms, mediated by several key signaling pathways (ErbB, p53, PI3K-Akt, VEGF and JAK-STAT), cytokines and hormones (estrogen, GnRH, relaxin, thyroid hormone, and prolactin). Elucidation of DE-miRNA predicted genes' specific roles, signal transduction pathways, and mechanisms, by mimics and inhibitors, which could perhaps offer diagnostic and therapeutic targets for infertility, cancer, and birth control.
Collapse
Affiliation(s)
- Vanmathy R Kasimanickam
- Center for Reproductive Biology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Ramanathan K Kasimanickam
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| |
Collapse
|
3
|
Corpuz-Hilsabeck M, Culty M. Impact of endocrine disrupting chemicals and pharmaceuticals on Sertoli cell development and functions. Front Endocrinol (Lausanne) 2023; 14:1095894. [PMID: 36793282 PMCID: PMC9922725 DOI: 10.3389/fendo.2023.1095894] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/04/2023] [Indexed: 02/01/2023] Open
Abstract
Sertoli cells play essential roles in male reproduction, from supporting fetal testis development to nurturing male germ cells from fetal life to adulthood. Dysregulating Sertoli cell functions can have lifelong adverse effects by jeopardizing early processes such as testis organogenesis, and long-lasting processes such as spermatogenesis. Exposure to endocrine disrupting chemicals (EDCs) is recognized as contributing to the rising incidence of male reproductive disorders and decreasing sperm counts and quality in humans. Some drugs also act as endocrine disruptors by exerting off-target effects on endocrine tissues. However, the mechanisms of toxicity of these compounds on male reproduction at doses compatible with human exposure are still not fully resolved, especially in the case of mixtures, which remain understudied. This review presents first an overview of the mechanisms regulating Sertoli cell development, maintenance, and functions, and then surveys what is known on the impact of EDCs and drugs on immature Sertoli cells, including individual compounds and mixtures, and pinpointing at knowledge gaps. Performing more studies on the impact of mixtures of EDCs and drugs at all ages is crucial to fully understand the adverse outcomes these chemicals may induce on the reproductive system.
Collapse
|
4
|
Wilharm A, Brigas HC, Sandrock I, Ribeiro M, Amado T, Reinhardt A, Demera A, Hoenicke L, Strowig T, Carvalho T, Prinz I, Ribot JC. Microbiota-dependent expansion of testicular IL-17-producing Vγ6 + γδ T cells upon puberty promotes local tissue immune surveillance. Mucosal Immunol 2021; 14:242-252. [PMID: 32733025 PMCID: PMC7790758 DOI: 10.1038/s41385-020-0330-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 07/01/2020] [Accepted: 07/14/2020] [Indexed: 02/04/2023]
Abstract
γδT cells represent the majority of lymphocytes in several mucosal tissues where they contribute to tissue homoeostasis, microbial defence and wound repair. Here we characterise a population of interleukin (IL) 17-producing γδ (γδ17) T cells that seed the testis of naive C57BL/6 mice, expand at puberty and persist throughout adulthood. We show that this population is foetal-derived and displays a T-cell receptor (TCR) repertoire highly biased towards Vγ6-containing rearrangements. These γδ17 cells were the major source of IL-17 in the testis, whereas αβ T cells mostly provided interferon (IFN)-γ in situ. Importantly, testicular γδ17 cell homoeostasis was strongly dependent on the microbiota and Toll-like receptor (TLR4)/IL-1α/IL-23 signalling. We further found that γδ17 cells contributed to tissue surveillance in a model of experimental orchitis induced by intra-testicular inoculation of Listeria monocytogenes, as Tcrδ-/- and Il17-/- infected mice displayed higher bacterial loads than wild-type (WT) controls and died 3 days after infection. Altogether, this study identified a previously unappreciated foetal-derived γδ17 cell subset that infiltrates the testis at steady state, expands upon puberty and plays a crucial role in local tissue immune surveillance.
Collapse
Affiliation(s)
- Anneke Wilharm
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Helena C Brigas
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Inga Sandrock
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Miguel Ribeiro
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Tiago Amado
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Annika Reinhardt
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Abdi Demera
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Lisa Hoenicke
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Till Strowig
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Tânia Carvalho
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Immo Prinz
- Institute of Immunology, Hannover Medical School, Hannover, Germany.
| | - Julie C Ribot
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
5
|
E4 Transcription Factor 1 (E4F1) Regulates Sertoli Cell Proliferation and Fertility in Mice. Animals (Basel) 2020; 10:ani10091691. [PMID: 32962114 PMCID: PMC7552733 DOI: 10.3390/ani10091691] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/13/2020] [Accepted: 09/16/2020] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Male fertility relies on the generation of functional sperm in seminiferous tubules of the testis. In mammals, Sertoli cells are the only somatic cells that directly interact with spermatogenic cells. Compelling evidences suggest that the number of Sertoli cells determines testis size and sperm output, however, molecular mechanisms regulating Sertoli cell proliferation and maturation are not well-understood. Using a Sertoli cell specific loss-of-function approach, here we showed that transcription factor E4F1 played an important role in murine Sertoli cell proliferation. Compared with their littermate control, E4f1 conditional knockout male mice sired a significantly low number of pups. E4f1 deletion resulted in reduced Sertoli cell number and testis size. Further analyses revealed that E4f1 deletion affected Sertoli cell proliferation in the neonatal testis and caused an increase in apoptosis of spermatogenic cells without affecting normal development of spermatogonia, meiotic and post-meiotic germ cells. These findings have shed new light on molecular controlling of spermatogenesis in mice and a similar mechanism likely exists in other animals. Abstract In the mammalian testes, Sertoli cells are the only somatic cells in the seminiferous tubules that provide structural, nutritional and regulatory support for developing spermatogenic cells. Sertoli cells only proliferate during the fetal and neonatal periods and enter a quiescent state after puberty. Functional evidences suggest that the size of Sertoli cell population determines sperm production and fertility. However, factors that direct Sertoli cell proliferation and maturation are not fully understood. Transcription factor E4F1 is a multifunctional protein that serves essential roles in cell fate decisions and because it interacts with pRB, a master regulator of Sertoli cell function, we hypothesized that E4F1 may have a functional role in Sertoli cells. E4f1 mRNA was present in murine testis and immunohistochemical staining confirmed that E4F1 was enriched in mature Sertoli cells. We generated a conditional knockout mouse model using Amh-cre and E4f1flox/flox lines to study E4F1 fucntion in Sertoli cells and the results showed that E4f1 deletion caused a significant reduction in testis size and fertility. Further analyses revealed that meiosis progression and spermiogenesis were normal, however, Sertoli cell proliferation was impaired and germ cell apoptosis was elevated in the testis of E4f1 conditional knockout mice. On the basis of these findings, we concluded that E4F1 was expressed in murine Sertoli cells and served important functions in regulating Sertoli cell proliferation and fertility.
Collapse
|
6
|
Azimzadeh M, Jelodar G. Alteration of testicular regulatory and functional molecules following long-time exposure to 900 MHz RFW emitted from BTS. Andrologia 2019; 51:e13372. [PMID: 31347712 DOI: 10.1111/and.13372] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 06/12/2019] [Accepted: 06/21/2019] [Indexed: 12/31/2022] Open
Abstract
The aim of this investigation was to evaluate changes in testosterone and some of the functional and regulatory molecules of testis such as P450scc, steroidogenic acute regulatory protein (StAR), tumour necrosis factor-α (TNF-α), interleukin-1α (IL-1α), interleukin-1β (IL-1β) and nerve growth factor (NGF) following exposure to 900 MHz radio frequency (RF). Thirty adult male Sprague Dawley rats (190 ± 20 g BW) were randomly classified in three equal groups, control (sham, without any exposure), short-time exposure (2 hr) (STE) and long-time exposure (4 hr) (LTE). The exposure was performed for 30 consecutive days. The testosterone level in both exposed groups was significantly less than control (p < .05). Level of TNF-α in both exposed groups was significantly greater than control (p < .05). IL-1α and NGF levels in LTE were significantly higher than the STE and control groups (p < .05). Level of IL-1β in LTE was significantly higher than control (p < .05). Expression of both P450scc and StAR mRNA was significantly down-regulated in both exposed groups compared to control (p < .05). Our results showed that RFW can affect testis and reproductive function through changes in factors, which are important during steroidogenesis, and also through changes in inflammatory factors, which regulate Leydig cell functions.
Collapse
Affiliation(s)
- Mansour Azimzadeh
- Department of Basic Science, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Gholamali Jelodar
- Department of Basic Science, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| |
Collapse
|
7
|
Neirijnck Y, Kühne F, Mayère C, Pavlova E, Sararols P, Foti M, Atanassova N, Nef S. Tumor Suppressor PTEN Regulates Negatively Sertoli Cell Proliferation, Testis Size, and Sperm Production In Vivo. Endocrinology 2019; 160:387-398. [PMID: 30576429 DOI: 10.1210/en.2018-00892] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 12/17/2018] [Indexed: 11/19/2022]
Abstract
The IGFs are the major intratesticular factors regulating immature Sertoli cell proliferation and are, therefore, critical to establish the magnitude of sperm production. However, the intratesticular source of IGF production and the downstream signaling pathway mediating IGF-dependent Sertoli cell proliferation remain unclear. Single-cell RNA sequencing on mouse embryonic testis revealed a robust expression of Igf1 and Igf2 in interstitial steroidogenic progenitors, suggesting that IGFs exert paracrine actions on immature Sertoli cells. To elucidate the intracellular signaling mechanism that underlies the proliferative effects of IGFs on immature Sertoli cells, we have generated mice with Sertoli cell-specific deletion of the Pten gene, a negative regulator of the phosphatidylinositol-3 kinase (PI3K)/AKT pathway, alone or together with the insulin receptor (Insr) and the IGF1 receptor (Igf1r). Although ablation of Pten appears dispensable for Sertoli cell proliferation and spermatogenesis, inactivation of Pten in the absence of Insr and Igf1r rescued the Sertoli cell proliferation rate during late fetal development, testis size, and sperm production. Overall, these findings suggest that IGFs secreted by interstitial progenitor cells act in a paracrine fashion to promote the proliferation of immature Sertoli cells through the IGF/PTEN/PI3K pathway.
Collapse
Affiliation(s)
- Yasmine Neirijnck
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Françoise Kühne
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Chloé Mayère
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Ekaterina Pavlova
- Institute of Experimental Morphology, Pathology and Anthropology with Museum, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Pauline Sararols
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Michelangelo Foti
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Nina Atanassova
- Institute of Experimental Morphology, Pathology and Anthropology with Museum, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Serge Nef
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
8
|
Siervo GEML, Ogo FM, Staurengo-Ferrari L, Anselmo-Franci JA, Cunha FQ, Cecchini R, Guarnier FA, Verri WA, Fernandes GSA. Sleep restriction during peripuberty unbalances sexual hormones and testicular cytokines in rats. Biol Reprod 2019; 100:112-122. [PMID: 30010983 DOI: 10.1093/biolre/ioy161] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 07/13/2018] [Indexed: 12/18/2022] Open
Abstract
Spermatogenesis and steroidogenesis are not fully established during puberty. Especially during this period, children and adolescents may be chronically sleep deprived due to early school hours and constant exposure to artificial light and interactive activities. We have previously shown that sleep restriction (SR) during peripuberty impairs sperm motility and has consequences on epididymal development in rats. Thus, this study aimed to evaluate the effect of SR during peripuberty on sexual hormones and its impact on testicular tissue. Rats were subjected to 18 h of SR per day for 21 days or were maintained as controls (C) in the same room. The circulating luteinizing hormone levels were decreased in SR rats without changes in the follicle stimulating hormone levels. Plasma and intratesticular testosterone and corticosterone in the SR group were increased in relation to C group. These alterations impair testicular tissue, with decreased IL-1β, IL-6, and TNFα levels in the testis and diminished seminiferous epithelium height and Sertoli cell number. SR also increased testicular lipid peroxidation with no alteration in antioxidant profiles. There were no significant changes in sperm parameters, seminiferous tubule diameter, histopathology, spermatogenesis kinetics, neutrophil and macrophage recruitment, and IL-10 concentration. Our results show that SR unbalances sexual hormones and testicular cytokines at a critical period of sexual maturation. These changes lead to lipid peroxidation in the testes and negatively influence the testicular tissue, as evidenced by diminished seminiferous epithelium height-with apoptosis of germinative cell-and Sertoli cell number.
Collapse
Affiliation(s)
- Gláucia E M L Siervo
- Department of Pathological Sciences, Biological Sciences Center, State University of Londrina, Londrina, Paraná, Brazil.,Department of General Biology, Biological Sciences Center, State University of Londrina, Londrina, Paraná, Brazil
| | - Fernanda M Ogo
- Department of Pathological Sciences, Biological Sciences Center, State University of Londrina, Londrina, Paraná, Brazil.,Department of General Biology, Biological Sciences Center, State University of Londrina, Londrina, Paraná, Brazil
| | - Larissa Staurengo-Ferrari
- Department of Pathological Sciences, Biological Sciences Center, State University of Londrina, Londrina, Paraná, Brazil
| | - Janete A Anselmo-Franci
- Department of Morphology, Stomatology and Physiology, Dental School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Fernando Q Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Rubens Cecchini
- Department of Pathological Sciences, Biological Sciences Center, State University of Londrina, Londrina, Paraná, Brazil
| | - Flávia A Guarnier
- Department of Pathological Sciences, Biological Sciences Center, State University of Londrina, Londrina, Paraná, Brazil
| | - Waldiceu A Verri
- Department of Pathological Sciences, Biological Sciences Center, State University of Londrina, Londrina, Paraná, Brazil
| | - Glaura S A Fernandes
- Department of General Biology, Biological Sciences Center, State University of Londrina, Londrina, Paraná, Brazil
| |
Collapse
|
9
|
Meroni SB, Galardo MN, Rindone G, Gorga A, Riera MF, Cigorraga SB. Molecular Mechanisms and Signaling Pathways Involved in Sertoli Cell Proliferation. Front Endocrinol (Lausanne) 2019; 10:224. [PMID: 31040821 PMCID: PMC6476933 DOI: 10.3389/fendo.2019.00224] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 03/21/2019] [Indexed: 12/16/2022] Open
Abstract
Sertoli cells are somatic cells present in seminiferous tubules which have essential roles in regulating spermatogenesis. Considering that each Sertoli cell is able to support a limited number of germ cells, the final number of Sertoli cells reached during the proliferative period determines sperm production capacity. Only immature Sertoli cells, which have not established the blood-testis barrier, proliferate. A number of hormonal cues regulate Sertoli cell proliferation. Among them, FSH, the insulin family of growth factors, activin, and cytokines action must be highlighted. It has been demonstrated that cAMP/PKA, ERK1/2, PI3K/Akt, and mTORC1/p70SK6 pathways are the main signal transduction pathways involved in Sertoli cell proliferation. Additionally, c-Myc and hypoxia inducible factor are transcription factors which participate in the induction by FSH of various genes of relevance in cell cycle progression. Cessation of proliferation is a pre-requisite to Sertoli cell maturation accompanied by the establishment of the blood-testis barrier. With respect to this barrier, the participation of androgens, estrogens, thyroid hormones, retinoic acid and opioids has been reported. Additionally, two central enzymes that are involved in sensing cell energy status have been associated with the suppression of Sertoli cell proliferation, namely AMPK and Sirtuin 1 (SIRT1). Among the molecular mechanisms involved in the cessation of proliferation and in the maturation of Sertoli cells, it is worth mentioning the up-regulation of the cell cycle inhibitors p21Cip1, p27Kip, and p19INK4, and of the gap junction protein connexin 43. A decrease in Sertoli cell proliferation due to administration of certain therapeutic drugs and exposure to xenobiotic agents before puberty has been experimentally demonstrated. This review focuses on the hormones, locally produced factors, signal transduction pathways, and molecular mechanisms controlling Sertoli cell proliferation and maturation. The comprehension of how the final number of Sertoli cells in adulthood is established constitutes a pre-requisite to understand the underlying causes responsible for the progressive decrease in sperm production that has been observed during the last 50 years in humans.
Collapse
|
10
|
Haverfield JT, Stanton PG, Loveland KL, Zahid H, Nicholls PK, Olcorn JS, Makanji Y, Itman CM, Simpson ER, Meachem SJ. Suppression of Sertoli cell tumour development during the first wave of spermatogenesis in inhibin α-deficient mice. Reprod Fertil Dev 2018; 29:609-620. [PMID: 26488911 DOI: 10.1071/rd15239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Accepted: 09/02/2015] [Indexed: 12/12/2022] Open
Abstract
A dynamic partnership between follicle-stimulating hormone (FSH) and activin is required for normal Sertoli cell development and fertility. Disruptions to this partnership trigger Sertoli cells to deviate from their normal developmental pathway, as observed in inhibin α-knockout (Inha-KO) mice, which feature Sertoli cell tumours in adulthood. Here, we identified the developmental windows by which adult Sertoli cell tumourigenesis is most FSH sensitive. FSH was suppressed for 7 days in Inha-KO mice and wild-type littermates during the 1st, 2nd or 4th week after birth and culled in the 5th week to assess the effect on adult Sertoli cell development. Tumour growth was profoundly reduced in adult Inha-KO mice in response to FSH suppression during Weeks 1 and 2, but not Week 4. Proliferative Sertoli cells were markedly reduced in adult Inha-KO mice following FSH suppression during Weeks 1, 2 or 4, resulting in levels similar to those in wild-type mice, with greatest effect observed at the 2 week time point. Apoptotic Sertoli cells increased in adult Inha-KO mice after FSH suppression during Week 4. In conclusion, acute FSH suppression during the 1st or 2nd week after birth in Inha-KO mice profoundly suppresses Sertoli cell tumour progression, probably by inhibiting proliferation in the adult, with early postnatal Sertoli cells being most sensitive to FSH action.
Collapse
Affiliation(s)
- Jenna T Haverfield
- Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Vic. 3168, Australia
| | - Peter G Stanton
- Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Vic. 3168, Australia
| | - Kate L Loveland
- Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Vic. 3168, Australia
| | - Heba Zahid
- Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Vic. 3168, Australia
| | - Peter K Nicholls
- Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Vic. 3168, Australia
| | - Justine S Olcorn
- Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Vic. 3168, Australia
| | - Yogeshwar Makanji
- Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Vic. 3168, Australia
| | - Catherine M Itman
- Priority Research Centres for Reproductive Science and Chemical Biology, School of Environmental and Life Sciences, Faculty of Science and Information Technology, University of Newcastle, University Drive, Callaghan, NSW 2308, Australia
| | - Evan R Simpson
- Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Vic. 3168, Australia
| | - Sarah J Meachem
- Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Vic. 3168, Australia
| |
Collapse
|
11
|
Zamani-Badi T, Nikzad H, Karimian M. IL-1RA
VNTR and IL-1α
4845G>T polymorphisms and risk of idiopathic male infertility in Iranian men: A case-control study and an in silico analysis. Andrologia 2018; 50:e13081. [DOI: 10.1111/and.13081] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 03/23/2018] [Accepted: 05/30/2018] [Indexed: 12/11/2022] Open
Affiliation(s)
- Tayyebeh Zamani-Badi
- Gametogenesis Research Center; Kashan University of Medical Sciences; Kashan Iran
| | - Hossein Nikzad
- Gametogenesis Research Center; Kashan University of Medical Sciences; Kashan Iran
| | - Mohammad Karimian
- Anatomical Sciences Research Center; Kashan University of Medical Sciences; Kashan Iran
| |
Collapse
|
12
|
Gautam M, Bhattacharya I, Rai U, Majumdar SS. Hormone induced differential transcriptome analysis of Sertoli cells during postnatal maturation of rat testes. PLoS One 2018; 13:e0191201. [PMID: 29342173 PMCID: PMC5771609 DOI: 10.1371/journal.pone.0191201] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Accepted: 12/30/2017] [Indexed: 11/18/2022] Open
Abstract
Sertoli cells (Sc) are unique somatic cells of testis that are the target of both FSH and testosterone (T) and regulate spermatogenesis. Although Sc of neonatal rat testes are exposed to high levels of FSH and T, robust differentiation of spermatogonial cells becomes conspicuous only after 11-days of postnatal age. We have demonstrated earlier that a developmental switch in terms of hormonal responsiveness occurs in rat Sc at around 12 days of postnatal age during the rapid transition of spermatogonia A to B. Therefore, such “functional maturation” of Sc, during pubertal development becomes prerequisite for the onset of spermatogenesis. However, a conspicuous difference in robust hormone (both T and FSH) induced gene expression during the different phases of Sc maturation restricts our understanding about molecular events necessary for the spermatogenic onset and maintenance. Here, using microarray technology, we for the first time have compared the differential transcriptional profile of Sc isolated and cultured from immature (5 days old), maturing (12 days old) and mature (60 days old) rat testes. Our data revealed that immature Sc express genes involved in cellular growth, metabolism, chemokines, cell division, MAPK and Wnt pathways, while mature Sc are more specialized expressing genes involved in glucose metabolism, phagocytosis, insulin signaling and cytoskeleton structuring. Taken together, this differential transcriptome data provide an important resource to reveal the molecular network of Sc maturation which is necessary to govern male germ cell differentiation, hence, will improve our current understanding of the etiology of some forms of idiopathic male infertility.
Collapse
Affiliation(s)
- Mukesh Gautam
- Department of Zoology, University of Delhi, Delhi, India
| | | | - Umesh Rai
- Department of Zoology, University of Delhi, Delhi, India
| | - Subeer S. Majumdar
- Cellular Endocrinology Laboratory, National Institute of Immunology, New Delhi, India
- National Institute of Animal Biotechnology, Hyderabad, India
- * E-mail:
| |
Collapse
|
13
|
Lee SLJ, Horsfield JA, Black MA, Rutherford K, Fisher A, Gemmell NJ. Histological and transcriptomic effects of 17α-methyltestosterone on zebrafish gonad development. BMC Genomics 2017; 18:557. [PMID: 28738802 PMCID: PMC5523153 DOI: 10.1186/s12864-017-3915-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 06/28/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Sex hormones play important roles in teleost ovarian and testicular development. In zebrafish, ovarian differentiation appears to be dictated by an oocyte-derived signal via Cyp19a1a aromatase-mediated estrogen production. Androgens and aromatase inhibitors can induce female-to-male sex reversal, however, the mechanisms underlying gonadal masculinisation are poorly understood. We used histological analyses together with RNA sequencing to characterise zebrafish gonadal transcriptomes and investigate the effects of 17α-methyltestosterone on gonadal differentiation. RESULTS At a morphological level, 17α-methyltestosterone (MT) masculinised gonads and accelerated spermatogenesis, and these changes were paralleled in masculinisation and de-feminisation of gonadal transcriptomes. MT treatment upregulated expression of genes involved in male sex determination and differentiation (amh, dmrt1, gsdf and wt1a) and those involved in 11-oxygenated androgen production (cyp11c1 and hsd11b2). It also repressed expression of ovarian development and folliculogenesis genes (bmp15, gdf9, figla, zp2.1 and zp3b). Furthermore, MT treatment altered epigenetic modification of histones in zebrafish gonads. Contrary to expectations, higher levels of cyp19a1a or foxl2 expression in control ovaries compared to MT-treated testes and control testes were not statistically significant during early gonad development (40 dpf). CONCLUSION Our study suggests that both androgen production and aromatase inhibition are important for androgen-induced gonadal masculinisation and natural testicular differentiation in zebrafish.
Collapse
Affiliation(s)
| | - Julia A. Horsfield
- Department of Pathology, University of Otago, Dunedin, Otago New Zealand
| | - Michael A. Black
- Department of Biochemistry, University of Otago, Dunedin, Otago New Zealand
| | - Kim Rutherford
- Department of Anatomy, University of Otago, Dunedin, Otago New Zealand
| | - Amanda Fisher
- Department of Pathology, University of Otago, Dunedin, Otago New Zealand
| | - Neil J. Gemmell
- Department of Anatomy, University of Otago, Dunedin, Otago New Zealand
| |
Collapse
|
14
|
Alves-Lopes JP, Söder O, Stukenborg JB. Testicular organoid generation by a novel in vitro three-layer gradient system. Biomaterials 2017; 130:76-89. [PMID: 28364632 DOI: 10.1016/j.biomaterials.2017.03.025] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 03/18/2017] [Accepted: 03/20/2017] [Indexed: 01/17/2023]
Abstract
A system that models the testicular microenvironment and spermatogonial stem-cell (SSC) niche in vitro has not been produced yet. Here, we developed and characterized a novel three-dimensional multilayer model, the Three-Layer Gradient System (3-LGS), which permits the generation of rat testicular organoids with a functional blood-testis barrier (BTB) and germ cell establishment and proliferation. The model is unique as regards the formation of cellular organizations that more closely represent the in vivo germ-to-somatic cell associations in vitro. Moreover, we also verified the roles of retinoic acid (RA), IL-1α, TNFα and RA inhibitors in germ cell maintenance and BTB organization in vitro. Treatment with RA was beneficial for germ cell maintenance, while IL-1α and TNFα were observed to impair the formation of testicular organoids and germ cell maintenance. Taking in account our characterization and validation results, we propose the 3-LGS as a new platform to investigate the SSC niche in vitro and to search for novel unknown factors involved in germ cell proliferation and differentiation. Moreover, we suggest that this model can be used in other scientific fields to study organogenesis and development by the generation of organoids.
Collapse
Affiliation(s)
- João Pedro Alves-Lopes
- Department of Women's and Children's Health, NORDFERTIL Research Lab Stockholm, Pediatric Endocrinology Unit, Q2:08, Karolinska Institutet and Karolinska University Hospital, SE-17176 Stockholm, Sweden.
| | - Olle Söder
- Department of Women's and Children's Health, NORDFERTIL Research Lab Stockholm, Pediatric Endocrinology Unit, Q2:08, Karolinska Institutet and Karolinska University Hospital, SE-17176 Stockholm, Sweden
| | - Jan-Bernd Stukenborg
- Department of Women's and Children's Health, NORDFERTIL Research Lab Stockholm, Pediatric Endocrinology Unit, Q2:08, Karolinska Institutet and Karolinska University Hospital, SE-17176 Stockholm, Sweden
| |
Collapse
|
15
|
Mital P, Kaur G, Bowlin B, Paniagua NJ, Korbutt GS, Dufour JM. Nondividing, postpubertal rat sertoli cells resumed proliferation after transplantation. Biol Reprod 2014; 90:13. [PMID: 24285718 DOI: 10.1095/biolreprod.113.110197] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Conventionally, it was believed that Sertoli cells (SC) stopped proliferating at puberty and became terminally differentiated quiescent cells. However, recent studies have challenged that dogma. In this study, we transplanted nondividing SC isolated from 23- to 27-day-old postpubertal rats transduced with a recombinant adenoviral vector (containing furin-modified human proinsulin cDNA) into diabetic severe combined immunodeficiency mice. Immunostaining the grafts for cell proliferation markers, proliferating cell nuclear antigen (PCNA) and MKI67, revealed that transplanted SC within the grafts were proliferating. Possible causes for resumption of proliferation of SC could be viral transduction, cell isolation and culture, higher abdominal temperature at the transplant site, and/or transplantation. To test for these possible causes, double- immunofluorescence staining was performed for GATA4 (SC marker) and MKI67. None of the SC were positive for MKI67 in tissue collected during SC isolation and culture or at higher temperature. However, nontransduced SC stained positive for MKI67 after transplantation into rats, suggesting viral transduction was not a key factor for induction of SC proliferation. Interestingly, resumption in proliferative ability of nondividing SC was temporary, as SC stopped proliferating within 14 days of transplantation and did not proliferate thereafter. Quantification of 5-bromo-2'-deoxyuridine-labeled SC demonstrated that 7%-9% of the total transplanted SC were proliferating in the grafts. These data indicate for the first time that nondividing SC resumed proliferation after transplantation and further validate previous findings that SC are not terminally differentiated. Hence, transplantation of SC could provide a useful model with which to study the regulation of SC proliferation in vivo.
Collapse
Affiliation(s)
- Payal Mital
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, Texas
| | | | | | | | | | | |
Collapse
|
16
|
Effects of testicular interstitial fluid on the proliferation of the mouse spermatogonial stem cells in vitro. ZYGOTE 2013; 22:395-403. [PMID: 23673083 DOI: 10.1017/s0967199413000142] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Spermatogenesis is a process in adult male mammals supported by spermatogonial stem cells (SSCs). The cultivation of SSCs has potential value, for example for the treatment of male infertility or spermatogonial transplantation. Testicular interstitial fluid was added to culture medium to a final concentration of 5, 10, 20, 30 or 40%, in order to investigate its effects on proliferation of mouse SSCs in vitro, Alkaline phosphatase (AKP) assay, reverse transcription polymerase chain reaction (RT-PCR) analysis and indirect immunofluorescence of cells were performed to identify SSCs, and the proliferation rate and diameters of the SSCs colonies were measured. The results showed that the optimal addition of testicular interstitial fluid to culture medium was 30%. When medium supplemented with 30% testicular interstitial fluid was used to culture mouse SSCs, the optimum proliferation rate and diameter of the cell colonies were 72.53% and 249 μm, respectively, after 8 days in culture, values that were significant higher than those found for other groups (P < 0.05). In conclusion, proliferation of mouse SSCs could be promoted significantly by supplementation of the culture medium with 30% testicular interstitial fluid. More research is needed to evaluate and understand the precise physiological role of testicular interstitial fluid during cultivation of SSCs.
Collapse
|
17
|
Pitetti JL, Calvel P, Zimmermann C, Conne B, Papaioannou MD, Aubry F, Cederroth CR, Urner F, Fumel B, Crausaz M, Docquier M, Herrera PL, Pralong F, Germond M, Guillou F, Jégou B, Nef S. An essential role for insulin and IGF1 receptors in regulating sertoli cell proliferation, testis size, and FSH action in mice. Mol Endocrinol 2013; 27:814-27. [PMID: 23518924 DOI: 10.1210/me.2012-1258] [Citation(s) in RCA: 169] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Testis size and sperm production are directly correlated to the total number of adult Sertoli cells (SCs). Although the establishment of an adequate number of SCs is crucial for future male fertility, the identification and characterization of the factors regulating SC survival, proliferation, and maturation remain incomplete. To investigate whether the IGF system is required for germ cell (GC) and SC development and function, we inactivated the insulin receptor (Insr), the IGF1 receptor (Igf1r), or both receptors specifically in the GC lineage or in SCs. Whereas ablation of insulin/IGF signaling appears dispensable for GCs and spermatogenesis, adult testes of mice lacking both Insr and Igf1r in SCs (SC-Insr;Igf1r) displayed a 75% reduction in testis size and daily sperm production as a result of a reduced proliferation rate of immature SCs during the late fetal and early neonatal testicular period. In addition, in vivo analyses revealed that FSH requires the insulin/IGF signaling pathway to mediate its proliferative effects on immature SCs. Collectively, these results emphasize the essential role played by growth factors of the insulin family in regulating the final number of SCs, testis size, and daily sperm output. They also indicate that the insulin/IGF signaling pathway is required for FSH-mediated SC proliferation.
Collapse
Affiliation(s)
- Jean-Luc Pitetti
- Department of Genetic Medicine and Development, National Center of Competence in Research, Frontiers in Genetics, University of Geneva, 1211 Geneva 4, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Alves MG, Rato L, Carvalho RA, Moreira PI, Socorro S, Oliveira PF. Hormonal control of Sertoli cell metabolism regulates spermatogenesis. Cell Mol Life Sci 2013; 70:777-93. [PMID: 23011766 PMCID: PMC11113727 DOI: 10.1007/s00018-012-1079-1] [Citation(s) in RCA: 129] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Revised: 06/11/2012] [Accepted: 06/26/2012] [Indexed: 11/30/2022]
Abstract
Hormonal regulation is essential to spermatogenesis. Sertoli cells (SCs) have functions that reach far beyond the physical support of germ cells, as they are responsible for creating the adequate ionic and metabolic environment for germ cell development. Thus, much attention has been given to the metabolic functioning of SCs. During spermatogenesis, germ cells are provided with suitable metabolic substrates, in a set of events mediated by SCs. Multiple signaling cascades regulate SC function and several of these signaling pathways are hormone-dependent and cell-specific. Within the seminiferous tubules, only SCs possess receptors for some hormones rendering them major targets for the hormonal signaling that regulates spermatogenesis. Although the mechanisms by which SCs fulfill their own and germ cells metabolic needs are mostly studied in vitro, SC metabolism is unquestionably a regulation point for germ cell development and the hormonal control of these processes is required for a normal spermatogenesis.
Collapse
Affiliation(s)
- Marco G. Alves
- CICS-UBI, Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Luís Rato
- CICS-UBI, Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Rui A. Carvalho
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
- Department of Life Sciences, Faculty of Science and Technology (FCTUC), University of Coimbra, 3004-517 Coimbra, Portugal
| | - Paula I. Moreira
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
- Institute of Physiology, Faculty of Medicine, University of Coimbra, 3000-354 Coimbra, Portugal
| | - Sílvia Socorro
- CICS-UBI, Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Pedro F. Oliveira
- CICS-UBI, Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| |
Collapse
|
19
|
Tarulli GA, Stanton PG, Meachem SJ. Is the adult Sertoli cell terminally differentiated? Biol Reprod 2012; 87:13, 1-11. [PMID: 22492971 DOI: 10.1095/biolreprod.111.095091] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
New data have challenged the convention that the adult Sertoli cell population is fixed and unmodifiable. The Sertoli cell has two distinct functions: 1) formation of the seminiferous cords and 2) provision of nutritional and structural support to developing germ cells. For these to occur successfully, Sertoli cells must undergo many maturational changes between fetal and adult life, the main switches occurring around puberty, including the loss of proliferative activity and the formation of the blood-testis barrier. Follicle-stimulating hormone plays a key role in promoting Sertoli cell proliferation, while thyroid hormone inhibits proliferative activity in early postnatal life. Together these regulate the Sertoli-germ cell complement and sperm output in adulthood. By puberty, the Sertoli cell population is considered to be stable and unmodifiable by hormones. But there is mounting evidence that the size of the adult Sertoli cell population and its maturational status is modifiable by hormones and that Sertoli cells can gain proliferative ability in the spermatogenically disrupted hamster and human model. This new information demonstrates that the adult Sertoli cell population, at least in the settings of testicular regression in the hamster and impaired fertility in humans in vivo and from mice and men in vitro, is not a terminally differentiated population. Data from the hamster now show that the adult Sertoli cell population size is regulated by hormones. This creates exciting prospects for basic and clinical research in testis biology. The potential to replenish an adult Sertoli-germ cell complement to normal in a setting of infertility may now be realized.
Collapse
Affiliation(s)
- Gerard A Tarulli
- Prince Henry's Institute of Medical Research, Clayton, Victoria, Australia
| | | | | |
Collapse
|
20
|
Lie PPY, Cheng CY, Mruk DD. The biology of interleukin-1: emerging concepts in the regulation of the actin cytoskeleton and cell junction dynamics. Cell Mol Life Sci 2012; 69:487-500. [PMID: 21744066 PMCID: PMC3297025 DOI: 10.1007/s00018-011-0760-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Revised: 06/14/2011] [Accepted: 06/17/2011] [Indexed: 01/28/2023]
Abstract
Interleukin (IL)-1 is a proinflammatory cytokine with important roles in innate immunity, as well as in normal tissue homeostasis. Interestingly, recent studies have also shown IL-1 to function in the dynamics of the actin cytoskeleton and cell junctions. For example, treatment of different epithelia with IL-1α often results in the restructuring of the actin network and cell junctions, thereby leading to junction disassembly. In this review, we highlight new and interesting findings that show IL-1 to be a critical player of restructuring events in the seminiferous epithelium of the testis during spermatogenesis.
Collapse
Affiliation(s)
- Pearl P. Y. Lie
- Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY 10065 USA
| | - C. Yan Cheng
- Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY 10065 USA
| | - Dolores D. Mruk
- Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY 10065 USA
| |
Collapse
|
21
|
Komsky A, Huleihel M, Ganaiem M, Kasterstein E, Komorovsky D, Bern O, Raziel A, Friedler S, Ron-El R, Strassburger D. Presence of IL-18 in testicular tissue of fertile and infertile men. Andrologia 2011; 44:1-8. [DOI: 10.1111/j.1439-0272.2010.01090.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
|
22
|
Izumi Y, Yamaguchi K, Ishikawa T, Ando M, Chiba K, Hashimoto H, Shiotani M, Fujisawa M. Molecular changes induced by bisphenol-A in rat Sertoli cell culture. Syst Biol Reprod Med 2011; 57:228-32. [DOI: 10.3109/19396368.2011.574248] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
23
|
Abu Elheija M, Dyomin V, Ganaiem M, Lunenfeld E, Vardy NS, Huleihel M. Distinct expression of interleukin-1α, interleukin-1β, and interleukin-1 receptor antagonist in testicular tissues and cells from human biopsies with normal and abnormal histology. J Interferon Cytokine Res 2011; 31:401-8. [PMID: 21235388 DOI: 10.1089/jir.2010.0059] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Cytokines are paracrine/autocrine growth factors known to affect testicular cell functions. The cellular origin and expression levels of interleukin-1 (IL-1) in human normal and pathological testicular biopsies are not yet clear. In the present study, we have investigated the levels and cellular origin of IL-1 family members [IL-1α, IL-β, and IL-1 receptor antagonist (IL-1ra)] in human testicular normal and abnormal biopsies with incomplete maturation arrest (IMA) or Sertoli only syndrome (SOS), using real-time polymerase chain reaction and immunohistochemical staining analysis. Our results show that the levels of IL-1α were higher in Leydig/interstitial cells of biopsies with IMA and SOS compared with normal. The levels of IL-1α in Sertoli cells of normal biopsies were higher than IMA and SOS. The mRNA levels of IL-1α were similar in all biopsies. IL-1β levels were higher in Leydig/interstitial cells of normal biopsies compared with Sertoli and germ cells. The levels of IL-1β were similar in testicular cells of all biopsies. However, the mRNA levels of IL-1β were significantly lower in SOS and IMA biopsies compared with normal. IL-1ra was expressed only in Leydig/interstitial cells, and their expression in normal biopsies was higher than in biopsies with IMA and SOS. The mRNA levels of IL-1ra were similar in all biopsies. Thus, it is possible to suggest the involvement of IL-1 system in the regulation of spermatogenesis and male infertility.
Collapse
Affiliation(s)
- Mahmoud Abu Elheija
- The Shraga Segal Department of Microbiology and Immunology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | | | | | | | | |
Collapse
|
24
|
Yang Y, Han C. GDNF stimulates the proliferation of cultured mouse immature Sertoli cells via its receptor subunit NCAM and ERK1/2 signaling pathway. BMC Cell Biol 2010; 11:78. [PMID: 20955573 PMCID: PMC2967512 DOI: 10.1186/1471-2121-11-78] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2010] [Accepted: 10/18/2010] [Indexed: 01/15/2023] Open
Abstract
Background The proliferation and final density of Sertoli cells in the testis are regulated by hormones and local factors. Glial cell line-derived neurotrophic factor (GDNF), a distantly related member of the transforming growth factor-β superfamily, and its receptor subunits GDNF family receptor alpha 1 (GFRα1), RET tyrosine kinase, and neural cell adhesion molecule (NCAM) have been reported to be expressed in the testis and involved in the regulation of proliferation of immature Sertoli cells (ISCs). However, the expression patterns of these receptor subunits and the downstream signaling pathways have not been addressed in ISCs. Results In the present study, we have reported that the proliferation of cultured ISCs was significantly enhanced by GDNF. The receptor subunits GFRα1 and NCAM but not RET were expressed in ISCs, and the stimulatory effect of GDNF on the proliferation of ISCs was significantly reduced by anti-NCAM antibody blocking or siRNA that specifically targets NCAM mRNA. Additionally, the ERK1/2 inhibitor, PD98059, completely abolished the mitogenic effect of GDNF on ISCs. Conclusions GDNF stimulates the proliferation of ISCs via its receptor subunit NCAM and the consequent activation of the ERK1/2 signaling pathway.
Collapse
Affiliation(s)
- Yongguang Yang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100080, China
| | | |
Collapse
|
25
|
17β-Estradiol Regulates Cultured Immature Boar Sertoli Cell Proliferation via the cAMP-ERK1/2 Pathway and the Estrogen Receptor β. ACTA ACUST UNITED AC 2010. [DOI: 10.1016/s1671-2927(09)60208-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
26
|
Ahmed EA, Rijbroek ADBV, Kal HB, Sadri-Ardekani H, Mizrak SC, Pelt AMV, Rooij DGD. Proliferative Activity In Vitro and DNA Repair Indicate that Adult Mouse and Human Sertoli Cells Are Not Terminally Differentiated, Quiescent Cells1. Biol Reprod 2009; 80:1084-91. [DOI: 10.1095/biolreprod.108.071662] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
27
|
Wang Y, Lui WY. Opposite effects of interleukin-1alpha and transforming growth factor-beta2 induce stage-specific regulation of junctional adhesion molecule-B gene in Sertoli cells. Endocrinology 2009; 150:2404-12. [PMID: 19164472 DOI: 10.1210/en.2008-1239] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In the mammalian testis, junctional adhesion molecule-B (JAM-B) is found at the blood-testis barrier between Sertoli cells and the apical ectoplasmic specializations between Sertoli and germ cells. The expression of JAM-B is tightly regulated to allow the transit of developing germ cells across the blood-testis barrier and the timely release of mature spermatids at stage VIII. In this study, the basal transcription of JAM-B in the mouse Sertoli cell line, MSC-1 cells, was examined. We found that the constitutive expression of JAM-B is carried out by the binding of specificity proteins (Sps), ETS domain transcription factor Elk-1 (Elk1), neuron-restrictive silencer factor (NRSF), and E2F transcription factor 3 (E2F3) to various cis-acting elements including TG interacting factor (TGIF), Elk-1, NRSF, and proximal Sp1 (pSp1) + E2F binding motifs. We also investigated the effects of two cytokines IL-1alpha and TGF-beta2 on JAM-B expression. IL-1alpha promotes JAM-B expression by facilitating the binding of Elk-1 to TGIF and pSp1 + E2F motifs in a p38-dependent manner, which leads to an additive effect on Sp1- and NRSF-mediated JAM-B transactivation. TGF-beta2 inhibits JAM-B transcription via the activation of mothers against decapentaplegic (Smad) proteins and activated Smads compete with specificity proteins (Sp1 and Sp3) for the TGIF motif, resulting in JAM-B repression. IL-1alpha and Smad3 expression have been reported to be stage specific. IL-1alpha is absent in the seminferous epithelium at stages VII-VIII, whereas a high level of nuclear Smad3 level is found at the same stages. This study shows for the first time that IL-1alpha and TGF-beta2 regulate JAM-B expression in an opposite manner, and in vitro data obtained herein provide some clues on how junctions are regulated in the testis.
Collapse
Affiliation(s)
- Yang Wang
- Division of Endocrinology, School of Biological Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| | | |
Collapse
|
28
|
Bagu ET, Gordon JR, Rawlings NC. Post-natal changes in testicular concentrations of interleukin-1 alpha and beta and interleukin-6 during sexual maturation in bulls. Reprod Domest Anim 2009; 45:336-41. [PMID: 19144032 DOI: 10.1111/j.1439-0531.2008.01318.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Based on observations in laboratory animals interleukins could be regulators of testicular development. The objects of this study were to see if interleukins (IL-1 and IL-6) are present in the developing bull testis and to establish the temporal patterns of concentrations of IL-1 and IL-6 in the bovine testis during development. Separate groups of six bull calves were castrated every 4 weeks from 5 to 33 weeks of age, and at 56 weeks of age. Mean testicular IL-1 alpha concentrations decreased (p < 0.01) from 5 to 9 weeks of age and 13 to 21 weeks of age. Mean testicular IL-1 beta concentrations decreased (p < 0.01) from 13 to 17 weeks of age and from 29 to 33 weeks of age. Mean IL-1 bioactivity increased from 13 to 17 weeks of age, decreased to 21 weeks, increased to 25 weeks, decreased to 29 weeks and decreased from 33 to 56 weeks of age (p < 0.05). Mean testicular IL-6 concentrations decreased (p < 0.05) from 9 to 13 weeks of age, increased (p < 0.05) to 21 weeks, decreased (p < 0.05) to 25 weeks, increased (p < 0.05) to 29 weeks and decreased (p < 0.01) to 56 weeks of age. In conclusion, testicular IL-1 alpha, IL-1 beta and IL-6 were found in the bovine testis and concentrations were age dependent. Testicular IL-1 alpha and IL-1 beta concentrations were highest in the early post-natal period; however, IL-1 bioactivity and IL-6 concentrations were greatest in the immediate pre-pubertal period. These findings suggest a functional role for interleukins in testicular development in the bull.
Collapse
Affiliation(s)
- E T Bagu
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | | | | |
Collapse
|
29
|
Abu Elhija M, Lunenfeld E, Huleihel M. ORIGINAL ARTICLE: LPS Increases the Expression Levels of IL-18, ICE and IL-18 R in Mouse Testes. Am J Reprod Immunol 2008; 60:361-71. [DOI: 10.1111/j.1600-0897.2008.00636.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
30
|
Mirza M, Petersen C, Nordqvist K, Sollerbrant K. Coxsackievirus and adenovirus receptor is up-regulated in migratory germ cells during passage of the blood-testis barrier. Endocrinology 2007; 148:5459-69. [PMID: 17690169 DOI: 10.1210/en.2007-0359] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The coxsackievirus and adenovirus receptor (CAR) is a cell adhesion molecule expressed in epithelial tight junctions and other cell-cell contacts. Using indirect immunofluorescence, quantitative RT-PCR, and Western blots, the expression and distribution of CAR in developing and adult testis are examined. CAR is highly expressed in both Sertoli and germ cells during perinatal and postnatal development, followed by a rapid down-regulation of both mRNA and protein levels. Interestingly, we find that CAR is a previously unknown downstream target for FSH because CAR mRNA levels were induced in primary cultures of FSH-stimulated Sertoli cells. In contrast to other epithelia, CAR is not a general component of tight junctions in the seminiferous epithelium, and Sertoli cells in the adult testis do not express CAR. Instead, CAR expression is stage dependent and specifically found in migratory germ cells. RT-PCR also demonstrated the presence of junctional adhesion molecule-like (JAML) in the testis. JAML was previously reported by others to form a functional complex with CAR regulating transepithelial migration of leukocytes. The expression of JAML in the testis suggests that a similar functional complex might be present during germ cell migration across the blood-testis barrier. Finally, an intermediate compartment occupied by CAR-positive, migrating germ cells and flanked by two occludin-containing junctions is identified. Together, these results implicate a function for CAR in testis morphogenesis and in migration of germ cells across the blood-testis barrier during spermatogenesis.
Collapse
Affiliation(s)
- Momina Mirza
- Ludwig Institute for Cancer Research, Stockholm Branch, Karolinska Institutet, Box 240, SE-17177, Stockholm, Sweden
| | | | | | | |
Collapse
|
31
|
Abstract
Sexual dimorphism is a term describing morphological differences between the sexes, but is often extended to include all differences observed between females and males. Sex differentiation in vertebrates is by definition sexually dimorphic and starts at the level of the sex chromosomes. In this review the sexual dimorphism of gonadal differentiation is discussed, with a focus on human development. In the embryo, the indifferent gonadal anlagen harbours four different cell lineages with bipotential fates dependent on the sex of the individual. The different paths taken by these cell lineages in male and female development are reviewed, along with other sexually dimorphic features of gonadal development. These include sex-determining genes, timing of events, dependence on germ cells, spatial organization of stromal cells, steroidogenic cells types, and other aspects.
Collapse
Affiliation(s)
- Olle Söder
- Department of Woman and Child Health, Paediatric Endocrinology Unit, Karolinska Institutet & University Hospital, SE-11130 Stockholm, Sweden.
| |
Collapse
|
32
|
Riera MF, Galardo MN, Pellizzari EH, Meroni SB, Cigorraga SB. Participation of phosphatidyl inositol 3-kinase/protein kinase B and ERK1/2 pathways in interleukin-1β stimulation of lactate production in Sertoli cells. Reproduction 2007; 133:763-73. [PMID: 17504920 DOI: 10.1530/rep.1.01091] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Interleukin-1β (IL1β ) belongs to a set of intratesticular regulators that provide the fine-tuning of cellular processes implicated in the maintenance of spermatogenesis. The aim of the present study was to analyze the signaling pathways that may participate in IL1β regulation of Sertoli cell function. Sertoli cell cultures from 20-day-old rat were used. Stimulation of the cultures with IL1β showed increments in phosphorylated protein kinase B (PKB), P70S6K, and ERK1/2 levels. A phosphatidyl inositol 3-kinase (PI3K) inhibitor (wortmannin (W)), a mammalian target of rapamycin inhibitor (rapamycin (R)), and a MEK inhibitor (PD98059 (PD)) were utilized to evaluate the participation of PI3K/PKB, P70S6K, and ERK1/2 pathways in the regulation of lactate production by IL1β . PD and W, but not R, decreased IL1β-stimulated lactate production. The participation of these pathways in the regulation of glucose uptake and lactate dehydrogenase (LDH) A mRNA levels by IL1β was also analyzed. It was observed that W decreased IL1β-stimulated glucose uptake, whereas PD and R did not modify it. On the other hand, PD decreased the stimulation of LDH A mRNA levels by IL1β , whereas W and R did not modify it. In summary, results presented herein demonstrate that IL1β stimulates PI3K/PKB-, P70S6K-, and ERK1/2-dependent pathways in rat Sertoli cells. Moreover, these results show that while IL1β utilizes the PI3K/PKB pathway to regulate glucose transport, it utilizes the ERK1/2 pathway to regulate LDH A mRNA levels. This study reveals that IL1β utilizes different signal transduction pathways to modify the biochemical steps that are important to regulate lactate production in rat Sertoli cells.
Collapse
Affiliation(s)
- María Fernanda Riera
- Centro de Investigaciones Endocrinológicas (CEDIE), Hospital de Niños Ricardo Gutiérrez, Gallo 1330, C1425EFD Buenos Aires, Argentina
| | | | | | | | | |
Collapse
|
33
|
Gerendai I, Banczerowski P, Csernus V, Halász B. Innervation and serotoninergic receptors of the testis interact with local action of interleukin-1beta on steroidogenesis. Auton Neurosci 2007; 131:21-7. [PMID: 16829209 DOI: 10.1016/j.autneu.2006.06.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2006] [Revised: 05/22/2006] [Accepted: 06/02/2006] [Indexed: 10/24/2022]
Abstract
Testosterone secretion by Leydig cells is affected by interleukin-1beta (IL-1beta). The aim of the present study was to investigate whether partial denervation of the testis or local administration of a serotonin (5-HT) receptor antagonist could alter the changes in testicular steoidogenesis induced by IL-1beta. Intratesticular administration of IL-1beta was combined with vasectomy or local injection of ketanserin (5-HT type 2 receptor antagonist) in immature hemicastrated rats and the effect of the interventions on testicular steroidogenesis was studied. One day after treatment with local injection of IL-1beta induced a significant rise in testosterone secretion that could be prevented by vasectomy (that also means transection of the inferior spermatic nerve). In a model in which neither IL-1beta nor ketanserin interfered with steroidogenesis, administration of the receptor antagonist just prior to IL-1beta treatment significantly reduced testosterone secretion. Data indicate interaction between testicular nerves and IL-1beta action and interaction between testicular 5-HT2 receptors and local effect of IL-1beta on testosterone secretion.
Collapse
Affiliation(s)
- Ida Gerendai
- Neuroendocrine Research Laboratory, Hungarian Academy of Sciences and Semmelweis University, Department of Human Morphology and Developmental Biology, H-1094 Budapest, Tuzoltó u. 58., Hungary.
| | | | | | | |
Collapse
|
34
|
Rozwadowska N, Fiszer D, Jedrzejczak P, Kosicki W, Kurpisz M. Interleukin-1 superfamily genes expression in normal or impaired human spermatogenesis. Genes Immun 2007; 8:100-7. [PMID: 17215863 DOI: 10.1038/sj.gene.6364356] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Interleukin-1 (IL-1) is a pleiotropic cytokine that may play a role in contributing to the specific immune environment of mammalian testis and in regulating cell differentiation. We have determined the transcription activity of the IL-1 gene family (using real-time polymerase chain reaction (PCR)) in two main functional testicular compartments (interstitial and intratubular ones), and in tissue homogenates obtained from patients with fertility disorders (spermatogenic arrest and testicular tumors). We observed the prominent expression of gene coding for IL-1 receptor antagonist (IL-1RA) in a purified fraction of gametogenic cells (normal gonad). Caspase-1 (ICE - IL-1beta-converting enzyme) was highly expressed (on mRNA level) in interstitial compartments as well in testicular tumors (immune enhancement?). In addition we found, that the activity of IL-1RA gene decreased along spermatogenic alteration in an inversely related manner with IL-1alpha (from normal gonad through spermatogenic arrest to Sertoli cell only syndrome). Therefore, the quotient value of IL-1alpha/IL-1RA could potentially serve as the diagnostic molecular probe for spermatogenesis assessment. The precise level of mRNA for IL-1-IL-18 cytokines and their receptors, and specifically of the receptor antagonist in immune privileged gonad, could be one of the main factors responsible for maintaining testicular homeostasis, thus enabling generation of the mature spermatozoa.
Collapse
Affiliation(s)
- N Rozwadowska
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | | | | | | | | |
Collapse
|
35
|
Colón E, Strand ML, Carlsson-Skwirut C, Wahlgren A, Svechnikov KV, Cohen P, Söder O. Anti-apoptotic factor humanin is expressed in the testis and prevents cell-death in leydig cells during the first wave of spermatogenesis. J Cell Physiol 2006; 208:373-85. [PMID: 16619233 DOI: 10.1002/jcp.20672] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Humanin (HN) is a 24 amino acids peptide with potent neuro-survival properties that protects against damage associated with Alzheimer's disease. In the present report, we have demonstrated by immunohistochemical analysis and Western blotting the pattern of expression of rat humanin (HNr) in the testis of 10- to 60-day-old rats. The Leydig cells of 10- and 40- day-old rats expressed this peptide at high levels; and in the testis of 60-day-old rats the expression of HNr expanded to include Leydig, endothelial, peritubular and germ cells. As monitored by Western blotting, HNr was released into the medium of cultures of Leydig cells isolated from 10-, 40-, and 60-days-old rats. HNr stimulated the incorporation of [(3)H]TdR into DNA of Leydig cells from 10-days-old rats, in a manner that indicated promotion of cell survival rather than an increase in the rate of cell multiplication. This peptide also enhanced steroidogenesis by cultured Leydig cells from 10- to 40-day-old rats both alone and synergistically with IGF-I. The expression of HNr in cultured Leydig cells increased in response to GH and IGF-I. In summary, we demonstrated here that HNr was expressed at all stages of maturation in the rat testis. This peptide promoted the survival of Leydig cells in culture and interacted with IGF-I to stimulate DNA synthesis and steroidogenesis. We propose that HNr is a novel testicular anti-apoptotic factor.
Collapse
Affiliation(s)
- Eugenia Colón
- Department of Woman and Child Health, Paediatric Endocrinology Unit, Astrid Lindgren Children's Hospital, Karolinska Institute and University Hospital, Stockholm, Sweden
| | | | | | | | | | | | | |
Collapse
|
36
|
Petersen C, Soder O. The sertoli cell--a hormonal target and 'super' nurse for germ cells that determines testicular size. HORMONE RESEARCH 2006; 66:153-61. [PMID: 16804315 DOI: 10.1159/000094142] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The somatic Sertoli cell plays an essential role in embryonic determination of male somatic sex and in spermatogenesis during adult life. One individual Sertoli cell supplies a clone of developing germ cells with nutrients and growth factors and it is well established that the number of Sertoli cells present is closely correlated to both testicular size and sperm output. Sertoli cells continue to proliferate and differentiate until the beginning of puberty, when they cease dividing and start nursing the germ cells. At this point in time, the future capacity of the testis for sperm production has thus been determined. Prior to puberty the Sertoli cells are immature and differ considerably with respect to morphology and biochemical activity from the mature cell. The several investigations that have focused on hormonal and paracrine regulation of the functions of the mature cell are reviewed here, but the mechanisms underlying the maturation and general maintenance of well-functioning Sertoli cells remain obscure. An alarming decline in male reproductive health has been observed in several Western countries during recent decades. Disturbance of Sertoli cell differentiation is thought to be involved in the pathogenesis of both a poor sperm count and testicular cancer. It is speculated that environmental agents that disrupt the estrogenic/androgenic balance in the testis may play a role in this connection.
Collapse
Affiliation(s)
- Cecilia Petersen
- Paediatric Endocrinology Unit, Department of Woman and Child Health, Astrid Lindgren Children's Hospital, Karolinska Institutet, Stockholm, Sweden.
| | | |
Collapse
|
37
|
Dahia CL, Rao AJ. Regulation of FSH receptor, PKIbeta, IL-6 and calcium mobilization: Possible mediators of differential action of FSH. Mol Cell Endocrinol 2006; 247:73-81. [PMID: 16406266 DOI: 10.1016/j.mce.2005.10.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2005] [Accepted: 10/31/2005] [Indexed: 11/19/2022]
Abstract
Sertoli cells support the development of germ cells by providing a microenvironment in the seminiferous tubules. FSH stimulates Sertoli cell proliferation only during neonatal period till day 18 in the immature rat whereas FSH regulates only functional parameters in the adult rat Sertoli cells. This suggests that FSH exerts differential action in immature and adult Sertoli cells. In an attempt to elucidate the mechanism by which FSH exerts the differential effects, we have carried out both in vivo and in vitro studies using Sertoli cells isolated from immature (7-10 days old) and adult (90 days old) rats. The differential role of FSH was studied at the receptor as well as at the signaling level. Monitoring the level of expression of FSH receptor by RTPCR and northern blot analysis revealed that the expression was more in immature Sertoli cells. Furthermore, it was found that FSH up (1.8-fold) regulates its receptor level only in the immature Sertoli cells and not in the adult. Results also revealed that PKIbeta and calcium, which are the downstream signaling molecules, are involved in FSH regulated Sertoli cells proliferation. It was also observed that FSH up (1.4-fold) regulates the levels of expression of IL-6 mRNA only in the immature rat Sertoli cells suggesting the possibility of its involvement in FSH regulated Sertoli cell proliferation.
Collapse
Affiliation(s)
- Chitra Lekha Dahia
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| | | |
Collapse
|
38
|
Fiorini C, Decrouy X, Defamie N, Segretain D, Pointis G. Opposite regulation of connexin33 and connexin43 by LPS and IL-1α in spermatogenesis. Am J Physiol Cell Physiol 2006; 290:C733-40. [PMID: 16236818 DOI: 10.1152/ajpcell.00106.2005] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The gap junction proteins, connexins (Cxs), are present in the testis, and among them, Cx43 play an essential role in spermatogenesis. In the present study, we investigated the testicular expression and regulation of another Cx, Cx33, previously described as a negative regulator of gap junction communication. Cx33 mRNA was present in testis and undetectable in heart, liver, ovary, and uterus. In the mature testis, Cx33 was specifically immunolocalized in the basal compartment of the seminiferous tubules, whereas Cx43 was present in both seminiferous tubule and interstitial compartments. During stages IX and X of spermatogenesis, characterized by Sertoli cell phagocytosis of residual bodies, Cx43 was poorly expressed within seminiferous tubules, while Cx33 signal was strong. To evaluate the role of phagocytosis in the control of Cx33 and Cx43 expression, the effect of LPS was analyzed in the Sertoli cell line 42GPA9. We show herein that phagocytosis activation by LPS concomitantly stimulated Cx33 and inhibited Cx43 mRNA levels. These effects appear to have been mediated through IL-1α, because the exposure of Sertoli cells to the IL-1 receptor antagonist partly reversed these effects. IL-1α enhanced and reduced, respectively, the levels of Cx33 and Cx43 mRNA in a time- and dose-dependent manner. These data reveal that Cx33 and Cx43 genes are controlled differently within the testis and suggest that these two Cxs may exert opposite and complementary effects on spermatogenesis.
Collapse
Affiliation(s)
- Celine Fiorini
- Faculté de Médecine, INSERM U 670, 28 Ave. de Valombrose, 06107 Nice cedex 2, France
| | | | | | | | | |
Collapse
|
39
|
Elhija MA, Lunenfeld E, Huleihel M. Induction of IL-1, in the Testes of Adult Mice, Following Subcutaneous Administration of Turpentine. Am J Reprod Immunol 2006; 55:136-44. [PMID: 16433833 DOI: 10.1111/j.1600-0897.2005.00344.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
PROBLEM Interleukin-1 family is present in the testicular homogenates and its cellular compartments. It has been suggested that IL-1 is involved in physiological and pathological functions of the testicular tissues. In the present study we examined the effect of acute mostly localized inflammation, using turpentine, on the expression levels of testicular IL-1 system. METHODS OF STUDY Mice were subcutaneously injected with steam-distilled turpentine or saline (control). Three hours to 10 days following the injection, mice were killed and testis and spleen were homogenized and examined for interleukin (IL)-1alpha, IL-1beta, and IL-1 receptor antagonist (IL-1ra) levels by enzyme-linked immunosorbent assay and polymerase chain reaction. RESULTS Subcutaneous injection of turpentine induced mice systemic inflammation, as indicated by significant increase in serum IL-1beta levels, and IL-1alpha, IL-1beta and IL-1ra in spleen homogenates. The levels of IL-1alpha, IL-1beta and IL-1ra were significantly induced in testicular homogenates of adult mice following subcutaneous injection of turpentine. The significant induction of testicular IL-1alpha was detected after 3-24 hr of turpentine injection and decreased later (after 3-10 days) to levels similar to the control. However, significant induction of testicular IL-1beta was detected only after 3-10 days of turpentine injection, and for testicular IL-1ra levels was detected after 3 hr to 6 days of turpentine injection, and after 10 days the levels were similar to the control. These results were also confirmed by mRNA expression of these factors. CONCLUSION Our results demonstrate for the first time the distant effect of acute localized inflammation on testicular IL-1 levels. Thus, transient inflammatory response to infectious/inflammatory agents at non-testicular sites that elicit systemic IL-1 response should be considered during clinical treatment as a possible factor of male infertility.
Collapse
Affiliation(s)
- Mahmoud Abu Elhija
- Department of Microbiology and Immunology and BGU Cancer Research Center, Beer-Sheva, Israel
| | | | | |
Collapse
|
40
|
Gerendai I, Banczerowski P, Csernus V. Interleukin 1-beta injected into the testis acutely stimulates and later attenuates testicular steroidogenesis of the immature rat. Endocrine 2005; 28:165-70. [PMID: 16388089 DOI: 10.1385/endo:28:2:165] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2005] [Revised: 08/17/2005] [Accepted: 08/23/2005] [Indexed: 11/11/2022]
Abstract
The effect of intratesticular administration of interleukin-1beta (IL-1beta) on steroidogenesis was studied in immature and adult rats. In 21-d-old animals local bilateral injection or unilateral administration of 0.1 microg/testis of IL-1beta to hemicastrates resulted in a significant increase in basal testosterone secretion in vitro and serum testosterone concentration one day posttreatment. Six days after treatment the cytokine induced opposite effect in animals with two testes in situ, i.e., it suppressed steroidogenesis. When IL-1beta was combined with hemi-castration, IL-1beta failed to alter the parameters studied. In adult animals subjected to bilateral treatment or to unilateral injection followed by hemicastration, IL-1beta in doses of 1.5 microg/testis or 15 microg/testis did not influence steroidogenesis and serum testosterone concentration. No change in serum LH and FSH concentration could be observed in any experimental group. The data suggest that the proinflammatory cytokine IL-1beta exerts a local action on testicular steroidogenesis, and the effect is age-dependent.
Collapse
Affiliation(s)
- Ida Gerendai
- Neuroendocrine Research Laboratory, Hungarian Academy of Sciences and Semmelweis University, Department of Human Morphology and Developmental Biology, H-1094 Budapest, Tuzoltó u. 58., Hungary.
| | | | | |
Collapse
|
41
|
Strand ML, Wahlgren A, Svechnikov K, Zetterström C, Setchell BP, Söder O. Interleukin-18 is expressed in rat testis and may promote germ cell growth. Mol Cell Endocrinol 2005; 240:64-73. [PMID: 16002206 DOI: 10.1016/j.mce.2005.05.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2005] [Revised: 05/16/2005] [Accepted: 05/25/2005] [Indexed: 10/25/2022]
Abstract
Although host-defence mechanisms, designed to preserve the integrity of the developing germ cells are operative in the testis, the components of this protective system have yet to be characterised in detail. Here, we report that the cytokine interleukin-18 (IL-18) is expressed in the rat testis and may contribute to these defences. Thus, analysis by RT-PCR and Western blotting revealed pronounced testicular expression of pro-IL-18 from postnatal day 5 and onwards. Expression of both IL-18 mRNA and protein was found to be localised to meiotic and post-meiotic germ cells as evaluated by in situ hybridisation and immunohistochemistry, respectively. The mRNA species coding for the IL-18 receptor and IL-1beta converting enzyme, which activates pro-IL-18, were also shown to be expressed by the seminiferous tubules. Recombinant IL-18 was seen to stimulate spermatogonial DNA synthesis in cultures of staged segments of rat seminiferous tubules, without influencing germ cell apoptosis. These results suggest that IL-18 may have host-protective and growth-promoting functions in the testis, but further investigations need to be done to confirm this.
Collapse
Affiliation(s)
- Mona-Lisa Strand
- Department of Woman and Child Health, Paediatric Endocrinology Unit Q2:08, Astrid Lindgren Children's Hospital, Karolinska Institute and University Hospital, Stockholm, Sweden.
| | | | | | | | | | | |
Collapse
|
42
|
Halberstadt C, Emerich DF, Gores P. Use of Sertoli cell transplants to provide local immunoprotection for tissue grafts. Expert Opin Biol Ther 2005; 4:813-25. [PMID: 15174964 DOI: 10.1517/14712598.4.6.813] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The recent success of allogeneic islet transplantation for the treatment of type I diabetes has renewed interest in cell therapy for diseases of secretory cell dysfunction. Unfortunately, widespread clinical use of cell transplantation is limited by tissue availability and the need for long-term immunosuppresion. Testicular Sertoli cells can confer local immunoprotection for co-transplanted cells and may provide a means of overcoming the obstacles associated with cell transplantation. Sertoli cell grafts protect islets in animal models of diabetes and can be transplanted into the brain to enhance regeneration and promote the survival of co-grafted tissues. This review describes the role that Sertoli cells normally play in testicular immunology, details the preclinical data using transplanted Sertoli cells in models of diabetes and Parkinson's disease and discusses some of the possible mechanisms involved in this phenomena, as well as the future of this technology.
Collapse
Affiliation(s)
- Craig Halberstadt
- Carolinas Medical Center, General Surgery Research, Cannon Research Center, P.O. Box 32861, Charlotte, NC 28232-2861, USA.
| | | | | |
Collapse
|
43
|
Chaudhary J, Sadler-Riggleman I, Ague JM, Skinner MK. The helix-loop-helix inhibitor of differentiation (ID) proteins induce post-mitotic terminally differentiated Sertoli cells to re-enter the cell cycle and proliferate. Biol Reprod 2005; 72:1205-17. [PMID: 15647457 DOI: 10.1095/biolreprod.104.035717] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Prior to puberty the Sertoli cells undergo active cell proliferation, and at the onset of puberty they become a terminally differentiated postmitotic cell population that support spermatogenesis. The molecular mechanisms involved in the postmitotic block of pubertal and adult Sertoli cells are unknown. The four known helix-loop-helix ID proteins (i.e., Id1, Id2, Id3, and Id4) are considered dominant negative regulators of cellular differentiation pathways and act as positive regulators of cellular proliferation. ID proteins are expressed at low levels by postpubertal Sertoli cells and are transiently induced by serum. The hypothesis tested was that ID proteins can induce a terminally differentiated postmitotic Sertoli cell to reenter the cell cycle if they are constitutively expressed. To test this hypothesis, ID1 and ID2 were stably integrated and individually overexpressed in postmitotic rat Sertoli cells. Overexpression of ID1 or ID2 allowed postmitotic Sertoli cells to reenter the cell cycle and undergo mitosis. The cells continued to proliferate even after 300 cell doublings. The functional markers of Sertoli cell differentiation such as transferrin, inhibin alpha, Sert1, and androgen binding protein (ABP) continued to be expressed by the proliferating Sertoli cells, but at lower levels. FSH receptor expression was lost in the proliferating Sertoli cell-Id lines. Some Sertoli cell genes, such as cyclic protein 2 (cathepsin L) and Sry-related HMG box protein-11 (Sox11) increase in expression. At no stage of proliferation did the cells exhibit senescence. The expression profile as determined with a microarray protocol of the Sertoli cell-Id lines suggested an overall increase in cell cycle genes and a decrease in growth inhibitory genes. These results demonstrate that overexpression of ID1 and ID2 genes in a postmitotic, terminally differentiated cell type have the capacity to induce reentry into the cell cycle. The observations are discussed in regards to potential future applications in model systems of terminally differentiated cell types such as neurons or myocytes.
Collapse
Affiliation(s)
- Jaideep Chaudhary
- Center for Reproductive Biology, School of Molecular Biosciences, Washington State University, Pullman,Washington 99164-4231, USA
| | | | | | | |
Collapse
|
44
|
Petersen C, Fröysa B, Söder O. Endotoxin and proinflammatory cytokines modulate Sertoli cell proliferation in vitro. J Reprod Immunol 2004; 61:13-30. [PMID: 15027475 DOI: 10.1016/j.jri.2003.10.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Sertoli cells play a key role in testicular function and their final number in the adult testis determines the capacity of germ cell production. Sertoli cell proliferation, stimulated by FSH and paracrine factors, occurs only in fetal and prepubertal life and may be an important target of pathogenic influences affecting testis development. We used a Sertoli cell proliferation assay to address the question whether if bacterial endotoxin (lipopolysaccharide; LPS) and proinflammatory cytokines could influence early postnatal Sertoli cell development. LPS and tumor necrosis factor-alpha (TNF-alpha) dose-dependently stimulated proliferation of primary cultures of isolated Sertoli cells from 8- to 9-day-old rats, assessed by (3)H-thymidine and BrdU incorporation. LPS also significantly increased the number of living cells in culture, measured by supravital staining. Interleukin-6 (IL-6) and interferon-gamma (IFN-gamma) had no direct effect on Sertoli cell growth, but were found to modulate FSH action. IL-6 increased, while IFN-gamma inhibited, FSH-induced Sertoli cell DNA-synthesis. We conclude that endotoxin and TNF-alpha are potent direct stimulators of Sertoli cell proliferation in vitro, and that IL-6 and IFN-gamma can modulate the mitogenic action of FSH on immature Sertoli cells. This may contribute to the pathogenesis of testicular damage after infections and inflammatory diseases in fetal and early postnatal life, with subsequent disturbance of adult germ cell production.
Collapse
Affiliation(s)
- Cecilia Petersen
- Department of Women and Child Health, Paediatric Endocrinology Unit, Astrid Lindgren Children's Hospital, Karolinska Institutet and Hospital, Stockholm, Sweden.
| | | | | |
Collapse
|
45
|
Lysiak JJ. The role of tumor necrosis factor-alpha and interleukin-1 in the mammalian testis and their involvement in testicular torsion and autoimmune orchitis. Reprod Biol Endocrinol 2004; 2:9. [PMID: 15012831 PMCID: PMC404472 DOI: 10.1186/1477-7827-2-9] [Citation(s) in RCA: 100] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2003] [Accepted: 03/10/2004] [Indexed: 12/04/2022] Open
Abstract
This review will focus the roles of TNF-alpha, IL-1 alpha, and IL-1 beta in the mammalian testis and in two testicular pathologies, testicular torsion and orchitis. TNF alpha in the testis is produced by round spermatids, pachytene spermatocytes, and testicular macrophages. The type 1 TNF receptor has been found on Sertoli and Leydig cells and numerous studies suggest a paracrine mode of action for TNF alpha in the normal testis. IL-1 alpha has been reported to be produced by Sertoli cells, testicular macrophages, and possibly postmeiotic germ cells. IL-1 receptors have been reported on Sertoli cells, Leydig cells, testicular macrophages, and germ cells suggesting both autocrine and paracrine functions. While these proinflammatory cytokines have important roles in normal testicular homeostasis, an elevation of their expression can lead to testicular dysfunctions. Testicular torsion is a clinical pathology with results in testicular ischemia and surgical intervention is often required for reperfusion. A pivotal role for IL-1beta in the pathology of testicular torsion has been recently described whereby an increase in IL-1beta production after reperfusion of the testis is correlated with the activation of the stress-related kinase, c-jun N-terminal kinase, and ultimately resulting in neutrophil recruitment to the testis and germ cell apoptosis. In autoimmune orchitis, on the other hand, TNF alpha produced by T-lymphocytes and macrophages of the testis has been implicated in the development and progression of the disease. Thus, both proinflammatory cytokines, TNF alpha and IL-1, have significant roles in normal testicular functions as well as in certain testicular pathologies.
Collapse
Affiliation(s)
- Jeffrey J Lysiak
- Department of Urology, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
46
|
Sultana T, Wahab-Wahlgren A, Assmus M, Parvinen M, Weber G, Söder O. Expression and regulation of the prointerleukin-1alpha processing enzymes calpain I and II in the rat testis. INTERNATIONAL JOURNAL OF ANDROLOGY 2003; 26:37-45. [PMID: 12534936 DOI: 10.1046/j.1365-2605.2003.00386.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Interleukin-1alpha (IL-1alpha) is constitutively expressed in an age- and stage-dependent manner by rat Sertoli cells. However, the mechanism of regulation of IL-1alpha is unclear in testis. We studied this regulation at the level of the enzyme calpain, a potential regulator that cleaves 32 kDa proIL-1alpha to produce mature 17 kDa IL-1alpha. Both calpain I and II were found to cleave recombinant rat testis 32proIL-1alpha in vitro. A temporary age-related increase in messenger RNA (mRNA) levels of calpain I was found in testis of 20- and 25-day-old rats, coinciding with important events of spermatogenesis and a gradual increase in IL-1alpha, while calpain II expression was constant. In response to lipopolysaccharide (LPS), calpain I protein levels were down-regulated in the seminiferous tubules, while calpain II was less affected. By contrast, the liver after LPS treatment showed up-regulated calpain I and II immunoreactive protein and reverse transcriptase chain reaction (RT-PCR) signal. Depleting Leydig cells by ethane 1,2-dimethane sulphonate treatment resulted in down-regulated calpain I mRNA and protein expression, whereas calpain II remained unchanged. In summary, there is a differential expression of calpain I and II under pathological conditions induced either by endotoxin stimuli or Leydig cell depletion, which may produce a differential effect on IL-1alpha processing.
Collapse
Affiliation(s)
- Taranum Sultana
- Department of Molecular Medicine, Tumor Biochemistry Unit, Karolinska Institutet and Hospital, Stockholm, Sweden.
| | | | | | | | | | | |
Collapse
|