1
|
Bartolini D, Arato I, Mancuso F, Giustarini D, Bellucci C, Vacca C, Aglietti MC, Stabile AM, Rossi R, Cruciani G, Rende M, Calafiore R, Luca G, Galli F. Melatonin modulates Nrf2 activity to protect porcine pre-pubertal Sertoli cells from the abnormal H 2 O 2 generation and reductive stress effects of cadmium. J Pineal Res 2022; 73:e12806. [PMID: 35524288 PMCID: PMC9539639 DOI: 10.1111/jpi.12806] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 05/04/2022] [Accepted: 05/05/2022] [Indexed: 11/27/2022]
Abstract
Melatonin (MLT) is a cytoprotective agent holding potential to prevent cadmium (Cd) toxicity and its impact in testicular function and fertility. In this study, we explored such potential in porcine pre-pubertal Sertoli cells (SCs). Cd toxicity resulted in impaired SC viability and function, abnormal cellular H2 O2 generation and efflux, and induction of reductive stress by the upregulation of Nrf2 expression and activity, cystine uptake and glutathione biosynthesis, glutathione-S-transferase P (GSTP) expression, and protein glutathionylation inhibition. Cd toxicity also stimulated the activity of cellular kinases (MAPK-ERK1/2 and Akt) and NFkB transcription factor, and cJun expression was increased. MLT produced a potent cytoprotective effect when co-administered with Cd to SCs; its efficacy and the molecular mechanism behind its cytoprotective function varied according to Cd concentrations. However, a significant restoration of cell viability and function, and of H2 O2 levels, was observed both at 5 and 10 μM Cd. Mechanistically, these effects of MLT were associated with a significant reduction of the Cd-induced activation of Nrf2 and GSTP expression at all Cd concentrations. CAT and MAPK-ERK1/2 activity upregulation was associated with these effects at 5 μM Cd, whereas glutathione biosynthesis and efflux were involved at 10 μM Cd together with an increased expression of the cystine transporter xCT, of cJun and Akt and NFkB activity. MLT protects SCs from Cd toxicity reducing its H2 O2 generation and reductive stress effects. A reduced activity of Nrf2 and the modulation of other molecular players of MLT signaling, provide a mechanistic rational for the cytoprotective effect of this molecule in SCs.
Collapse
Affiliation(s)
- Desirée Bartolini
- Department of Pharmaceutical SciencesUniversity of PerugiaPerugiaItaly
- Department of Medicine and Surgery, Section of Human, Clinical and Forensic AnatomyUniversity of PerugiaPerugiaItaly
| | - Iva Arato
- Department of Medicine and SurgeryUniversity of PerugiaPerugiaItaly
| | | | - Daniela Giustarini
- Department of Biotechnology, Chemistry and PharmacyUniversity of SienaSienaItaly
| | - Catia Bellucci
- Department of Medicine and SurgeryUniversity of PerugiaPerugiaItaly
| | - Carmine Vacca
- Department of Chemistry, Biology and BiotechnologyUniversity of PerugiaPerugiaItaly
| | | | - Anna Maria Stabile
- Department of Medicine and Surgery, Section of Human, Clinical and Forensic AnatomyUniversity of PerugiaPerugiaItaly
| | - Ranieri Rossi
- Department of Biotechnology, Chemistry and PharmacyUniversity of SienaSienaItaly
| | - Gabriele Cruciani
- Department of Chemistry, Biology and BiotechnologyUniversity of PerugiaPerugiaItaly
| | - Mario Rende
- Department of Medicine and Surgery, Section of Human, Clinical and Forensic AnatomyUniversity of PerugiaPerugiaItaly
| | - Riccardo Calafiore
- Department of Medicine and SurgeryUniversity of PerugiaPerugiaItaly
- Department of Medicine and Surgery, International Biotechnological Center for Endocrine, Metabolic and Embryo‐Reproductive Translational Research (CIRTEMER)University of PerugiaPerugiaItaly
| | - Giovanni Luca
- Department of Medicine and SurgeryUniversity of PerugiaPerugiaItaly
- Division of Medical Andrology and Endocrinology of ReproductionSaint Mary HospitalTerniItaly
- Department of Medicine and Surgery, International Biotechnological Center for Endocrine, Metabolic and Embryo‐Reproductive Translational Research (CIRTEMER)University of PerugiaPerugiaItaly
| | - Francesco Galli
- Department of Pharmaceutical SciencesUniversity of PerugiaPerugiaItaly
| |
Collapse
|
2
|
Cannarella R, Mancuso F, Arato I, Lilli C, Bellucci C, Gargaro M, Curto R, Aglietti MC, La Vignera S, Condorelli RA, Luca G, Calogero AE. Sperm-carried IGF2 downregulated the expression of mitogens produced by Sertoli cells: A paracrine mechanism for regulating spermatogenesis? Front Endocrinol (Lausanne) 2022; 13:1010796. [PMID: 36523595 PMCID: PMC9744929 DOI: 10.3389/fendo.2022.1010796] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 11/07/2022] [Indexed: 11/30/2022] Open
Abstract
INTRODUCTION Insulin-like growth factor 2 (IGF2) mRNA has been found in human and mouse spermatozoa. It is currently unknown whether the IGF2 protein is expressed in human spermatozoa and, if so, its possible role in the cross-talk between germ and Sertoli cells (SCs) during spermatogenesis. METHODS To accomplish this, we analyzed sperm samples from four consecutive Caucasian men. Furthermore, to understand its role during the spermatogenetic process, porcine SCs were incubated with increasing concentrations (0.33, 3.33, and 10 ng/mL) of recombinant human IGF2 (rhIGF2) for 48 hours. Subsequently, the experiments were repeated by pre-incubating SCs with the non-competitive insulin-like growth factor 1 receptor (IGF1R) inhibitor NVP-AEW541. The following outcomes were evaluated: 1) Gene expression of the glial cell-line derived neurotrophic factor (GDNF), fibroblast growth factor 2 (FGF2), and stem cell factor (SCF) mitogens; 2) gene and protein expression of follicle-stimulating hormone receptor (FSHR), anti-Müllerian hormone (AMH), and inhibin B; 3) SC proliferation. RESULTS We found that the IGF2 protein was present in each of the sperm samples. IGF2 appeared as a cytoplasmic protein localized in the equatorial and post-acrosomal segment and with a varying degree of expression in each cell. In SCs, IGF2 significantly downregulated GDNF gene expression in a concentration-dependent manner. FGF2 and SCF were downregulated only by the highest concentration of IGF2. Similarly, IGF2 downregulated the FSHR gene and FSHR, AMH, and inhibin B protein expression. Finally, IGF2 significantly suppressed the SC proliferation rate. All these findings were reversed by pre-incubation with NVP-AEW541, suggesting an effect mediated by the interaction of IGF2 with the IGFR. CONCLUSION In conclusion, sperm IGF2 seems to downregulate the expression of mitogens, which are known to be physiologically released by the SCs to promote gonocyte proliferation and spermatogonial fate adoption. These findings suggest the presence of paracrine regulatory mechanisms acting on the seminiferous epithelium during spermatogenesis, by which germ cells can influence the amount of mitogens released by the SCs, their sensitivity to FSH, and their rate of proliferation.
Collapse
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
- Glickman Urological & Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
- *Correspondence: Rossella Cannarella,
| | - Francesca Mancuso
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Iva Arato
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Cinzia Lilli
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Catia Bellucci
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Marco Gargaro
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Roberto Curto
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Maria C. Aglietti
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Rosita A. Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Giovani Luca
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Aldo E. Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| |
Collapse
|
3
|
Xia Q, Cui G, Fan Y, Wang X, Hu G, Wang L, Luo X, Yang L, Cai Q, Xu K, Guo W, Gao M, Li Y, Wu J, Li W, Chen J, Qi H, Peng G, Yao H. RNA helicase DDX5 acts as a critical regulator for survival of neonatal mouse gonocytes. Cell Prolif 2021; 54:e13000. [PMID: 33666296 PMCID: PMC8088469 DOI: 10.1111/cpr.13000] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/11/2021] [Accepted: 01/14/2021] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES Mammalian spermatogenesis is a biological process of male gamete formation. Gonocytes are the only precursors of spermatogonial stem cells (SSCs) which develop into mature spermatozoa. DDX5 is one of DEAD-box RNA helicases and expresses in male germ cells, suggesting that Ddx5 plays important functions during spermatogenesis. Here, we explore the functions of Ddx5 in regulating the specification of gonocytes. MATERIALS AND METHODS Germ cell-specific Ddx5 knockout (Ddx5-/- ) mice were generated. The morphology of testes and epididymides and fertility in both wild-type and Ddx5-/- mice were analysed. Single-cell RNA sequencing (scRNA-seq) was used to profile the transcriptome in testes from wild-type and Ddx5-/- mice at postnatal day (P) 2. Dysregulated genes were validated by single-cell qRT-PCR and immunofluorescent staining. RESULTS In male mice, Ddx5 was expressed in germ cells at different stages of development. Germ cell-specific Ddx5 knockout adult male mice were sterile due to completely devoid of germ cells. Male germ cells gradually disappeared in Ddx5-/- mice from E18.5 to P6. Single-cell transcriptome analysis showed that genes involved in cell cycle and glial cell line-derived neurotrophic factor (GDNF) pathway were significantly decreased in Ddx5-deficient gonocytes. Notably, Ddx5 ablation impeded the proliferation of gonocytes. CONCLUSIONS Our study reveals the critical roles of Ddx5 in fate determination of gonocytes, offering a novel insight into the pathogenesis of male sterility.
Collapse
|
4
|
Arato I, Ceccarelli V, Mancuso F, Bellucci C, Lilli C, Ferolla P, Perruccio K, D'Arpino A, Aglietti MC, Calafiore R, Cameron DF, Calvitti M, Baroni T, Vecchini A, Luca G. Effect of EPA on Neonatal Pig Sertoli Cells " In Vitro": A Possible Treatment to Help Maintain Fertility in Pre-Pubertal Boys Undergoing Treatment With Gonado-Toxic Therapies. Front Endocrinol (Lausanne) 2021; 12:694796. [PMID: 34093450 PMCID: PMC8174840 DOI: 10.3389/fendo.2021.694796] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 05/05/2021] [Indexed: 01/15/2023] Open
Abstract
The incidence of cancer in pre-pubertal boys has significantly increased and, it has been recognized that the gonado-toxic effect of the cancer treatments may lead to infertility. Here, we have evaluated the effects on porcine neonatal Sertoli cells (SCs) of three commonly used chemotherapy drugs; cisplatin, 4-Hydroperoxycyclophosphamide and doxorubicin. All three drugs induced a statistical reduction of 5-hydroxymethylcytosine in comparison with the control group, performed by Immunofluorescence Analysis. The gene and protein expression levels of GDNF, were significantly down-regulated after treatment to all three chemotherapy drugs comparison with the control group. Specifically, differences in the mRNA levels of GDNF were: 0,8200 ± 0,0440, 0,6400 ± 0,0140, 0,4400 ± 0,0130 fold change at 0.33, 1.66, and 3.33μM cisplatin concentrations, respectively (**p < 0.01 at 0.33 and 1.66 μM vs SCs and ***p < 0.001 at 3.33μM vs SCs); 0,6000 ± 0,0340, 0,4200 ± 0,0130 fold change at 50 and 100 μM of 4-Hydroperoxycyclophosphamide concentrations, respectively (**p < 0.01 at both these concentrations vs SCs); 0,7000 ± 0,0340, 0,6200 ± 0,0240, 0,4000 ± 0,0230 fold change at 0.1, 0.2 and 1 µM doxorubicin concentrations, respectively (**p < 0.01 at 0.1 and 0.2 μM vs SCs and ***p < 0.001 at 1 μM vs SCs). Differences in the protein expression levels of GDNF were: 0,7400 ± 0,0340, 0,2000 ± 0,0240, 0,0400 ± 0,0230 A.U. at 0.33, 1.66, and 3.33μM cisplatin concentrations, respectively (**p < 0.01 at both these concentrations vs SCs); 0,7300 ± 0,0340, 0,4000 ± 0,0130 A.U. at 50 and 100 μM of 4- Hydroperoxycyclophosphamide concentrations, respectively (**p < 0.01 at both these concentrations vs SCs); 0,6200 ± 0,0340, 0,4000 ± 0,0240, 0,3800 ± 0,0230 A.U. at 0.l, 0.2 and 1 µM doxorubicin concentrations, respectively (**p < 0.01 at 0.1 and 0.2 μM vs SCs and ***p < 0.001 at 1 μM vs SCs). Furthermore, we have demonstrated the protective effect of eicosapentaenoic acid on SCs only at the highest concentration of cisplatin, resulting in an increase in both gene and protein expression levels of GDNF (1,3400 ± 0,0280 fold change; **p < 0.01 vs SCs); and of AMH and inhibin B that were significantly recovered with values comparable to the control group. Results from this study, offers the opportunity to develop future therapeutic strategies for male fertility management, especially in pre-pubertal boys.
Collapse
Affiliation(s)
- Iva Arato
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Francesca Mancuso
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Catia Bellucci
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Cinzia Lilli
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Piero Ferolla
- Department of Medical Oncology, Multidisciplinary Neuroendocrine Tumours (NET) Group, Umbria Regional Cancer Network and University of Perugia, Perugia, Italy
| | - Katia Perruccio
- Pediatric Oncology Hematology, Department of Mother and Child Health, Perugia, Italy
| | | | | | - Riccardo Calafiore
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- International Biotechnological Center for Endocrine, Metabolic and Embryo-Reproductive Translational Research (CIRTEMER), Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Don F Cameron
- Morisani College of Medicine FL, University of South Florida, Tampa, FL, United States
| | - Mario Calvitti
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Tiziano Baroni
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Alba Vecchini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Giovanni Luca
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- International Biotechnological Center for Endocrine, Metabolic and Embryo-Reproductive Translational Research (CIRTEMER), Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- Division of Medical Andrology and Endocrinology of Reproduction, Saint Mary Hospital, Terni, Italy
| |
Collapse
|
5
|
Tenuta M, Carlomagno F, Cangiano B, Kanakis G, Pozza C, Sbardella E, Isidori AM, Krausz C, Gianfrilli D. Somatotropic-Testicular Axis: A crosstalk between GH/IGF-I and gonadal hormones during development, transition, and adult age. Andrology 2020; 9:168-184. [PMID: 33021069 DOI: 10.1111/andr.12918] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/31/2020] [Accepted: 09/28/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND The hypothalamic-pituitary-gonadal (HPG) and hypothalamic-pituitary-somatotropic (HPS) axes are strongly interconnected. Interactions between these axes are complex and poorly understood. These interactions are characterized by redundancies in reciprocal influences at each level of regulation and the combination of endocrine and paracrine effects that change during development. OBJECTIVES To comprehensively review the crosstalk between the HPG and HPS axes and related pathological and clinical aspects during various life stages of male subjects. MATERIALS AND METHODS A thorough search of publications available in PubMed was performed using proper keywords. RESULTS Molecular studies confirmed the expressions of growth hormone (GH) and insulin-like growth factor-I (IGF-I) receptors on the HPG axis and reproductive organs, indicating a possible interaction between HPS and HPG axes at various levels. Insulin growth factors participate in sexual differentiation during fetal development, indicating that normal HPS axis activity is required for proper testicular development. IGF-I contributes to correct testicular position during minipuberty, determines linear growth during childhood, and promotes puberty onset and pace through gonadotropin-releasing hormone activation. IGF-I levels are high during transition age, even when linear growth is almost complete, suggesting its role in reproductive tract maturation. Patients with GH deficiency (GHD) and insensitivity (GHI) exhibit delayed puberty and impaired genital development; replacement therapy in such patients induces proper pubertal development. In adults, few studies have suggested that lower IGF-I levels are associated with impaired sperm parameters. DISCUSSION AND CONCLUSION The role of GH-IGF-I in testicular development remains largely unexplored. However, it is important to evaluate gonadic development in children with GHD. Additionally, HPS axis function should be evaluated in children with urogenital malformation or gonadal development alterations. Correct diagnosis and prompt therapeutic intervention are needed for healthy puberty, attainment of complete gonadal development during transition age, and fertility potential in adulthood.
Collapse
Affiliation(s)
- Marta Tenuta
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | | | - Biagio Cangiano
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - George Kanakis
- Athens Naval and Veterans Affairs Hospital, Athens, Greece
| | - Carlotta Pozza
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Emilia Sbardella
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Andrea M Isidori
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Csilla Krausz
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | | |
Collapse
|
6
|
Boguslawska J, Kryst P, Poletajew S, Piekielko-Witkowska A. TGF-β and microRNA Interplay in Genitourinary Cancers. Cells 2019; 8:E1619. [PMID: 31842336 PMCID: PMC6952810 DOI: 10.3390/cells8121619] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 12/12/2022] Open
Abstract
Genitourinary cancers (GCs) include a large group of different types of tumors localizing to the kidney, bladder, prostate, testis, and penis. Despite highly divergent molecular patterns, most GCs share commonly disturbed signaling pathways that involve the activity of TGF-β (transforming growth factor beta). TGF-β is a pleiotropic cytokine that regulates key cancer-related molecular and cellular processes, including proliferation, migration, invasion, apoptosis, and chemoresistance. The understanding of the mechanisms of TGF-β actions in cancer is hindered by the "TGF-β paradox" in which early stages of cancerogenic process are suppressed by TGF-β while advanced stages are stimulated by its activity. A growing body of evidence suggests that these paradoxical TGF-β actions could result from the interplay with microRNAs: Short, non-coding RNAs that regulate gene expression by binding to target transcripts and inducing mRNA degradation or inhibition of translation. Here, we discuss the current knowledge of TGF-β signaling in GCs. Importantly, TGF-β signaling and microRNA-mediated regulation of gene expression often act in complicated feedback circuits that involve other crucial regulators of cancer progression (e.g., androgen receptor). Furthermore, recently published in vitro and in vivo studies clearly indicate that the interplay between microRNAs and the TGF-β signaling pathway offers new potential treatment options for GC patients.
Collapse
Affiliation(s)
- Joanna Boguslawska
- Department of Biochemistry and Molecular Biology, Centre of Postgraduate Medical Education; 01-813 Warsaw, Poland;
| | - Piotr Kryst
- II Department of Urology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland; (P.K.); (S.P.)
| | - Slawomir Poletajew
- II Department of Urology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland; (P.K.); (S.P.)
| | | |
Collapse
|
7
|
Zhang J, Yao D, Song Y, Pan Y, Zhu L, Bai Y, Xu Y, Zhang J, Shao C. Fractionated irradiation of right thorax induces abscopal damage on testes leading to decline in fertility. Sci Rep 2019; 9:15221. [PMID: 31645625 PMCID: PMC6811594 DOI: 10.1038/s41598-019-51772-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 09/27/2019] [Indexed: 01/16/2023] Open
Abstract
Radiation-induced abscopal effect (RIAE) may influence radiotherapy efficiency. However, it is unknown whether RIAE triggers abnormal genetic consequence. We present a novel evidence that, when mice were given fractionated irradiation on right thorax, the ultrastructure of blood-testis barrier was damaged in company with apoptosis induction in testes, and the sperm number and vitality were drastically decreased so that both the fertility and the survival of their offspring were reduced. Protein microarray assay and hormone detection showed that some cytokines especially TNF-α, TGF-β and estradiol in the serum of irradiated mice increased to higher levels in consistent with abscopal damage, and this conditioned serum had toxic effect on TM4 cells in vitro. When the mice were fed with cimetidine, the above abscopal responses were significantly attenuated. This study demonstrates in the first time that the thoracic irradiation (Th-IR) induces structural and functional damage in the distal testes and further cause fertility decline of irradiated male mice, which may have important implications in the strategy development of radiotherapy in avoiding abnormal genetic consequence.
Collapse
Affiliation(s)
- Junling Zhang
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Dan Yao
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yimeng Song
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yan Pan
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Lin Zhu
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yang Bai
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yanwu Xu
- Department of Biochemistry, College of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianghong Zhang
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Chunlin Shao
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
8
|
Özbek M, Hitit M, Öztop M, Beyaz F, Ergün E, Ergün L. Spatiotemporal expression patterns of natriuretic peptides in rat testis and epididymis during postnatal development. Andrologia 2019; 51:e13387. [PMID: 31661170 DOI: 10.1111/and.13387] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 05/28/2019] [Accepted: 07/11/2019] [Indexed: 01/19/2023] Open
Abstract
Natriuretic peptide (NP) family is composed of atrial, brain and C-type NP (NPPA, NPPB and NPPC). Here, we aimed to investigate NP expression in testis and epididymis during postnatal development. NPPA expression was observed in gonocytes at prepubertal period but in only spermatocytes in pachytene and leptotene/zygotene stage at pubertal period. In prepubertal and pubertal periods, we detected NPPB expression in only Leydig cells. However, NPPC expression was detected in all of the gonocytes and Sertoli cells, spermatocytes and some interstitial cells in prepubertal and pubertal periods. In postpubertal and mature periods, NPPA and NPPB staining were detected in Leydig cells, elongated and round spermatids but not in spermatogonia and spermatocytes. However, we observed NPPC expression in all cells of the seminiferous tubules and Leydig cells in the postpubertal and mature periods. Epididymal epithelium showed intense NPPC expression during postnatal period but weak NPPA and NPPB expression in prepubertal and pubertal periods. The expression of three NPs in the testis significantly increased after puberty. In conclusion, puberty had a significant effect on NP expression in testis. Unlike NPPA and NPPB, expression of NPPC in all cells of the seminiferous tubule suggests that NPPC is effective in each step of spermatogenesis.
Collapse
Affiliation(s)
- Mehmet Özbek
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Mehmet Akif Ersoy University, Burdur, Turkey
| | - Mustafa Hitit
- Department of Genetics, Faculty of Veterinary Medicine, Kastamonu University, Kastamonu, Turkey
| | - Mustafa Öztop
- Department of Biology, Faculty of Arts and Sciences, Mehmet Akif Ersoy University, Burdur, Turkey
| | - Feyzullah Beyaz
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Erciyes University, Kayseri, Turkey
| | - Emel Ergün
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Ankara University, Ankara, Turkey
| | - Levent Ergün
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Ankara University, Ankara, Turkey
| |
Collapse
|
9
|
Mora Rodríguez JA, Porchia LM, Camargo F, López-Bayghen E. The use of insulin-like growth factor 1 improved the parameters of the seminogram in a patient with severe oligoasthenoteratozoospermia. SAGE Open Med Case Rep 2019; 7:2050313X19834154. [PMID: 30858973 PMCID: PMC6404043 DOI: 10.1177/2050313x19834154] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 02/05/2019] [Indexed: 11/26/2022] Open
Abstract
Male patients suffering from oligoasthenoteratozoospermia typically failed to
achieve pregnancy, even with assisted reproductive technologies. Growth hormone
and insulin-like growth factor 1 have been shown to regulate sperm quality
parameters; therefore, the insulin-like growth factor 1 supplement could improve
sperm parameters. Here, we determine the effect insulin-like growth factor 1 has
on sperm parameters in a patient suffering from oligoasthenoteratozoospermia. A
47-year-old male was administered once a day 1.5 IU of insulin-like growth
factor 1 by intradermal injection for 2 months. Seminogram analysis was
performed before and after. Treatment with insulin-like growth factor 1 resulted
in a 15.5-fold improvement in sperm concentration (1.1 × 106 vs 18.3
× 106 per mL), 71.4% change in volume (0.7 vs 1.2 mL), increased
progressive motility (2% vs 43%), and the total volume of sperm with progressive
motility (0% vs 23.6%). Here, we show that administering a daily dose of
insulin-like growth factor 1 can improve sperm quality parameters.
Collapse
Affiliation(s)
- José Arturo Mora Rodríguez
- Laboratorio de Fertilización In Vitro and Laboratorio de Investigación y Diagnóstico Molecular, Instituto de Infertilidad y Genética México SC, Ingenes, México City, México
| | - Leonardo M Porchia
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), México City, México
| | - Felipe Camargo
- Laboratorio de Fertilización In Vitro and Laboratorio de Investigación y Diagnóstico Molecular, Instituto de Infertilidad y Genética México SC, Ingenes, México City, México
| | - Esther López-Bayghen
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), México City, México
| |
Collapse
|
10
|
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: 157] [Impact Index Per Article: 26.2] [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
|
11
|
Curley M, Milne L, Smith S, Atanassova N, Rebourcet D, Darbey A, Hadoke PWF, Wells S, Smith LB. Leukemia Inhibitory Factor-Receptor is Dispensable for Prenatal Testis Development but is Required in Sertoli cells for Normal Spermatogenesis in Mice. Sci Rep 2018; 8:11532. [PMID: 30068994 PMCID: PMC6070476 DOI: 10.1038/s41598-018-30011-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 07/19/2018] [Indexed: 12/14/2022] Open
Abstract
Leukemia inhibitory factor (LIF), a pleiotropic cytokine belonging to the interleukin-6 family, is most often noted for its role in maintaining the balance between stem cell proliferation and differentiation. In rodents, LIF is expressed in both the fetal and adult testis; with the peritubular myoid (PTM) cells thought to be the main site of production. Given their anatomical location, LIF produced by PTM cells may act both on intratubular and interstitial cells to influence spermatogenesis and steroidogenesis respectively. Indeed, the leukemia inhibitory factor receptor (LIFR) is expressed in germ cells, Sertoli cells, Leydig cells, PTM cells and testicular macrophages, suggesting that LIF signalling via LIFR may be a key paracrine regulator of testicular function. However, a precise role(s) for testicular LIFR-signalling in vivo has not been established. To this end, we generated and characterised the testicular phenotype of mice lacking LIFR either in germ cells, Sertoli cells or both, to identify a role for LIFR-signalling in testicular development/function. Our analyses reveal that LIFR is dispensable in germ cells for normal spermatogenesis. However, Sertoli cell LIFR ablation results in a degenerative phenotype, characterised by abnormal germ cell loss, sperm stasis, seminiferous tubule distention and subsequent atrophy of the seminiferous tubules.
Collapse
Affiliation(s)
- Michael Curley
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, United Kingdom
| | - Laura Milne
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, United Kingdom
| | - Sarah Smith
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, United Kingdom
| | - Nina Atanassova
- Institute of Experimental Morphology, Pathology and Anthropology with Museum, Bulgarian Academy of Sciences, 1113, Sofia, Bulgaria
| | - Diane Rebourcet
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, United Kingdom
| | - Annalucia Darbey
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, United Kingdom
| | - Patrick W F Hadoke
- The British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, EH16 4TJ, United Kingdom
| | - Sara Wells
- Mary Lyons Centre, MRC Harwell, Harwell Campus, Oxfordshire, OX11 ORD, United Kingdom
| | - Lee B Smith
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, United Kingdom. .,School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW, 2308, Australia.
| |
Collapse
|
12
|
Pampanini V, Germani D, Puglianiello A, Stukenborg JB, Reda A, Savchuk I, Kjartansdóttir KR, Cianfarani S, Söder O. Impact of uteroplacental insufficiency on postnatal rat male gonad. J Endocrinol 2017; 232:247-257. [PMID: 27885054 PMCID: PMC5184772 DOI: 10.1530/joe-16-0418] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 11/24/2016] [Indexed: 11/30/2022]
Abstract
Prenatal events such as intrauterine growth restriction can affect gonadal development of the offspring and have an impact on reproductive health. To investigate the effects of intrauterine growth restriction induced by uterine artery ligation on the postnatal rat testis. Pregnant rats underwent uterine artery ligation at day 19 of gestation. Offspring were killed at 5, 20 and 40 days post-partum (dpp). At killing, one gonad was snap-frozen in liquid nitrogen and processed for RNA and steroid extraction. The other gonad was formalin-fixed for histology. Gene expression was analyzed by TaqMan Low-Density Array. Intratesticular testosterone, estradiol and serum gonadotrophins were measured. Thirty genes were dysregulated in intrauterine growth-restricted rats compared to controls, among which markers of Sertoli cell and Leydig cell function, cell metabolism and growth factors. Testis weights were significantly reduced at 5 and 20 dpp in intrauterine growth-restricted rats and caught-up by 40 dpp Accordingly, Sertoli cell number was significantly lower in 5 dpp intrauterine growth-restricted rats. At 20 dpp, intratesticular testosterone was significantly increased in intrauterine growth-restricted rats, whereas serum gonadotrophins were unchanged. IUGR altered the gene expression in the rat testes up to peripubertal age and reduced testis size and Sertoli cell number in neonatal age. Multiple mechanisms encompassing genetic changes and steroid production by the testis may be involved in the catch-up growth phase that restored testis size by 40 dpp Permanent consequences on organ function and gamete integrity cannot be excluded and deserve further investigations.
Collapse
Affiliation(s)
- Valentina Pampanini
- Department of Women's and Children's HealthPediatric Endocrinology Unit, Q2:08, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Daniela Germani
- Department of Systems MedicineTor Vergata University, Rome, Italy
| | | | - Jan-Bernd Stukenborg
- Department of Women's and Children's HealthPediatric Endocrinology Unit, Q2:08, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Ahmed Reda
- Department of Women's and Children's HealthPediatric Endocrinology Unit, Q2:08, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Iuliia Savchuk
- Department of Women's and Children's HealthPediatric Endocrinology Unit, Q2:08, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Kristín Rós Kjartansdóttir
- Department of Women's and Children's HealthPediatric Endocrinology Unit, Q2:08, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Stefano Cianfarani
- Department of Women's and Children's HealthPediatric Endocrinology Unit, Q2:08, Karolinska Institutet and University Hospital, Stockholm, Sweden
- Dipartimento Pediatrico Universitario Ospedaliero 'Bambino Gesù' Children's Hospital - Tor Vergata UniversityRome, Italy
| | - Olle Söder
- Department of Women's and Children's HealthPediatric Endocrinology Unit, Q2:08, Karolinska Institutet and University Hospital, Stockholm, Sweden
| |
Collapse
|
13
|
Okada R, Hara T, Sato T, Kojima N, Nishina Y. The mechanism and control of Jagged1 expression in Sertoli cells. Regen Ther 2016; 3:75-81. [PMID: 31245476 PMCID: PMC6581826 DOI: 10.1016/j.reth.2016.02.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 02/08/2016] [Accepted: 02/15/2016] [Indexed: 01/25/2023] Open
Abstract
The regulation of Sertoli cells by some hormones and signaling factors is important for normal spermatogenesis. Notch signaling is considered to be necessary for normal spermatogenesis in mouse. In this study, we revealed two new facts about Sertoli cells by western blotting experiments on different types of primary cells and microdissected tubules. The first is that Sertoli cells express the Jagged1 ligand in mice testes. The second is that the expression level of Jagged1 oscillates in the seminiferous epithelial cycle. Therefore, we inferred that Jagged1 in Sertoli cells contributes to the Notch signaling involved in spermatogenesis. Furthermore, we examined the regulation of Jagged1 expression and found that Jagged1 expression was suppressed by cAMP signaling and was promoted by TNF-α signaling in Sertoli cells. When cAMP and TNF-α were simultaneously added to Sertoli cells, Jagged1 expression was suppressed. Therefore, cAMP signaling dominates Jagged1 expression over TNF-α signaling. These results suggest that cAMP signaling may cause the periodicity of Jagged1 expression in the seminiferous epithelial cycle, and controlling Jagged1 expression by adding TNF-α or cAMP may contribute to normal spermatogenesis in vitro. Jagged1 was expressed in Sertoli cells in mouse testes. The expression of Jagged1 oscillated in the seminiferous epithelial cycle. The expression of Jagged1 in Sertoli cells was upregulated by TNF-α and downregulated by cAMP.
Collapse
Affiliation(s)
- Ryu Okada
- Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama 236-0027, Japan
| | - Taro Hara
- Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama 236-0027, Japan
| | - Tomomi Sato
- Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama 236-0027, Japan
| | - Nobuhiko Kojima
- Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama 236-0027, Japan
| | - Yukio Nishina
- Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama 236-0027, Japan
| |
Collapse
|
14
|
Wang S, Wang X, Wu Y, Han C. IGF-1R signaling is essential for the proliferation of cultured mouse spermatogonial stem cells by promoting the G2/M progression of the cell cycle. Stem Cells Dev 2014; 24:471-83. [PMID: 25356638 DOI: 10.1089/scd.2014.0376] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Culture of mouse spermatogonial stem cells (mSSCs) contributes to understanding the mechanisms of mammalian spermatogenesis. Several key growth factors such as GDNF and FGF2 have been known to be essential for the proliferation of cultured mSSCs. However, additional factors regulating SSC proliferation remain to be identified. In this study, we report that IGF-1R signaling is required for the proliferation of cultured mSSCs by promoting the G2/M progression of the cell cycle. IGF-1 and its receptor IGF-1R are expressed in cultured mSSCs as well as in isolated Sertoli cells and interstitial cells. Blockage of IGF-1R signaling either by knockdown of IGF-1R or by the IGF-1R-specific inhibitor picropodophyllin (PPP) significantly reduced the proliferation of mSSCs, increased their apoptosis, and impaired their stem cell activity in an insulin-independent manner. PPP treatment of mSSCs blocked the G2/M progression. In contrast, both GDNF withdrawal and FGF2 signaling blockade decreased the entry of mSSCs into their S phases. Consistently, IGF-1 promoted the G2/M progression of thymidine-treated mSSCs, which were arrested at G1/S boundary synchronously; while GDNF and/or FGF2 stimulated their entry into the S phase. Moreover, IGF-1 activated the phosphorylation of AKT but not that of ERK1/2 in mSSCs. These results indicate that IGF-1R signaling stimulates the proliferation of mSSCs using a distinct mechanism from those by GDNF and FGF2, and will contribute to the establishment of a chemically defined culture system.
Collapse
Affiliation(s)
- Si Wang
- 1 State Key Laboratory of Reproductive Biology, Institute of Zoology , Chinese Academy of Sciences, Beijing, China
| | | | | | | |
Collapse
|
15
|
Yoon M, Jiang J, Chung KH, Roser JF. Immunolocalization of insulin-like growth factor-I (IGF-I) and its receptors (IGF-IR) in the equine epididymis. J Reprod Dev 2014; 61:30-4. [PMID: 25311540 PMCID: PMC4354228 DOI: 10.1262/jrd.2014-097] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Insulin-like growth factor plays a paracrine/autocrine role in regulating testicular function in the stallion, but its presence in the equine epididymis remains unknown. The aim of this study was to test the hypothesis that insulin-like growth factor-I (IGF-I) and IGF-I receptor (IGF-IR) are localized in the caput, corpus, and cauda of the epididymis in an age-dependent manner. Immediately after castration, epididymal tissue was fixed, paraffin-embedded, and processed for immunohistochemistry (IHC). Western blot was also performed using equine epididymal extracts to verify the specificity of the antibodies against IGF-I and IGF-IR. Immunolabeling of IGF-I was observed in the cytoplasm of principal and basal cells in the caput, corpus, and cauda at the pre-pubertal (3–7 months), pubertal (12–18 months), post-pubertal (2–4 years), and adult stages (4.5–8 years). Immunolabeling of IGF-IR was observed in the cytoplasm of principal cells in all regions of the epididymis in
each age group. Immunolabeling of IGF-IR was also detected in the cytoplasm of basal cells from animals of all ages. Bands observed by Western blot corresponded to the molecular weights of IGF-I and IGF-IR, ~23 kDa and 95 kDa, respectively. These results suggest that IGF-I might function as an autocrine and/or paracrine factor during the development, maintenance and/or secretions of the stallion epididymis.
Collapse
|
16
|
Transforming growth factor-β1 signaling represses testicular steroidogenesis through cross-talk with orphan nuclear receptor Nur77. PLoS One 2014; 9:e104812. [PMID: 25140527 PMCID: PMC4139307 DOI: 10.1371/journal.pone.0104812] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 07/15/2014] [Indexed: 11/25/2022] Open
Abstract
Transforming growth factor- β1 (TGF-β1) has been reported to inhibit luteinizing hormone (LH) mediated-steroidogenesis in testicular Leydig cells. However, the mechanism by which TGF-β1 controls the steroidogenesis in Leydig cells is not well understood. Here, we investigated the possibility that TGF-β1 represses steroidogenesis through cross-talk with the orphan nuclear receptor Nur77. Nur77, which is induced by LH/cAMP signaling, is one of major transcription factors that regulate the expression of steroidogenic genes in Leydig cells. TGF-β1 signaling inhibited cAMP-induced testosterone production and the expression of steroidogenic genes such as P450c17, StAR and 3β-HSD in mouse Leydig cells. Further, TGF-β1/ALK5 signaling repressed cAMP-induced and Nur77-activated promoter activity of steroidogenic genes. In addition, TGF-β1/ALK5-activated Smad3 repressed Nur77 transactivation of steroidogenic gene promoters by interfering with Nur77 binding to DNA. In primary Leydig cells isolated from Tgfbr2flox/flox Cyp17iCre mice, TGF-β1-mediated repression of cAMP-induced steroidogenic gene expression was significantly less than that in primary Leydig cells from Tgfbr2flox/flox mice. Taken together, these results suggest that TGF-β1/ALK5/Smad3 signaling represses the expression of steroidogenic genes via the suppression of Nur77 transactivation in testicular Leydig cells. These findings may provide a molecular mechanism involved in the TGF-β1-mediated repression of testicular steroidogenesis.
Collapse
|
17
|
Sokanovic SJ, Baburski AZ, Janjic MM, Stojkov NJ, Bjelic MM, Lalosevic D, Andric SA, Stojilkovic SS, Kostic TS. The opposing roles of nitric oxide and cGMP in the age-associated decline in rat testicular steroidogenesis. Endocrinology 2013; 154:3914-24. [PMID: 23885018 PMCID: PMC3776867 DOI: 10.1210/en.2013-1307] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The molecular mechanism of the aging-associated dysfunction of Leydig cells (LCs) is complex and poorly understood. In this study, we analyzed the contribution of nitric oxide (NO) and cGMP signaling to the age-dependent decline in LC function. Significant (>50%) decreases in serum, intratesticular, and LC androgens in aging rats (15-24 months) were accompanied by a proportional increase in NO production, an up-regulation of cGMP levels, and the expression of soluble guanylyl cyclase-1B and protein kinase G1 in LCs. In contrast, LC cAMP levels decreased with age, most likely reflecting the up-regulation of cAMP-specific phosphodiesterase expression. Moreover, the expression of genes encoding enzymes responsible for cholesterol transport and its conversion to T were reduced. Exposing LCs from aged animals to NO further increased cGMP levels and decreased cAMP and androgen production, whereas the addition of cell-permeable 8-bromoguanosine-cGMP alone had the opposite effect. In vivo inhibition of cGMP-specific phosphodiesterase-5 for 3 and 6 months in aged rats led to a partial restoration of androgens, NO, and cyclic nucleotide levels, as well as the expression of steroidogenic and NO/cGMP signaling genes. These results indicate that a progressive increase in NO production contributes to the age-dependent decrease in steroidogenesis in a cGMP-independent manner, whereas the sustained elevation in cGMP levels significantly slows the decline in LC function.
Collapse
Affiliation(s)
- Srdjan J Sokanovic
- PhD, Reproductive Endocrinology and Signaling Group, University of Novi Sad, Novi Sad 21000, Serbia.
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Galardo MN, Riera MF, Regueira M, Pellizzari EH, Cigorraga SB, Meroni SB. Different signal transduction pathways elicited by basic fibroblast growth factor and interleukin 1β regulate CREB phosphorylation in Sertoli cells. J Endocrinol Invest 2013; 36:331-8. [PMID: 22932066 DOI: 10.3275/8582] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND AND AIM Basic fibroblast growth factor (bFGF) and interleukin 1β (IL1β) belong to the set of intratesticular regulators that provide for the fine-tuning of processes implicated in the maintenance of spermatogenesis. The aim of this study was to investigate if bFGF and IL1β activate CREB, what signaling pathways may be participating and the possible relationship between CREB activation and the regulation of Sertoli cell function. METHODS Twenty-day-old rat Sertoli cell cultures were used. RESULTS Cultures stimulated with bFGF and IL1β produced a time-dependent increment in phosphorylated CREB levels that reached maximal values in 5- and 15-minute incubations respectively. MEK inhibitors--PD98059 and U0126--blocked the effect of bFGF on phosphorylated CREB while a p38-MAPK inhibitor--SB203580--blocked the effect of IL1β on phosphorylated CREB. A possible correlation between CREB regulation and two Sertoli cell-differentiated functions, Ldh A and transferrin expression, was explored. PD98059 blocked the ability of bFGF to stimulate Ldh A expression and SB203580 blocked the ability of IL1β to stimulate Ldh A expression and LDH activity. Concerning transferrin, PD98059 and U0126 were able to inhibit the ability of bFGF to stimulate its secre tion. On the contrary, SB203580 was unable to block IL1β induced increase in transferrin secretion suggesting that the p38-MAPK pathway does not participate in the mechanism of action of the cytokine to regulate transferrin. CONCLUSIONS The results presented herein suggest that CREB is stimulated in response to bFGF and IL1β through p42/p44-MAPK and p38-MAPK pathways and that this transcription factor may be partially responsible for the regulation of Sertoli cell function.
Collapse
Affiliation(s)
- M N Galardo
- Centro de Investigaciones Endocrinológicas, CEDIE-CONICET, Hospital de Niños Ricardo Gutiérrez, Gallo 1330, C1425EDF Buenos Aires, Argentina
| | | | | | | | | | | |
Collapse
|
19
|
Babu SR, Sadhnani MD, Swarna M, Padmavathi P, Reddy PP. Evaluation of FSH, LH and testosterone levels in different subgroups of infertile males. Indian J Clin Biochem 2012; 19:45-9. [PMID: 23105425 DOI: 10.1007/bf02872388] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Gonadotropins (FSH, LH) and testosterone are the prime regulators of germ cell development. Abnormal spermatogenesis is often associated with altered serum gonadotropins and testosterone. FSH, LH and testosterone levels were estimated in 96 infertile men of whom 35 were azoospermic, 35 were oligozoospermic, 11 were with varicocele and 15 were with histopathological abnormalities like hypospermatogenesis, spermatid arrest and sertoli-cell only syndrome. Results showed statistically significant (p<0.05), increase in the mean FSH and LH levels in all the infertile males studied when compared with the fertile controls (n=35). However, there is no significant difference in the mean levels of testosterone between the infertile and fertile men.
Collapse
Affiliation(s)
- S Ramesh Babu
- Institute of Genetics & Hospital for Genetic Diseases, Osmania University, Begumpet, 500 016 Hyderabad
| | | | | | | | | |
Collapse
|
20
|
Gettler LT, McKenna JJ, McDade TW, Agustin SS, Kuzawa CW. Does cosleeping contribute to lower testosterone levels in fathers? Evidence from the Philippines. PLoS One 2012; 7:e41559. [PMID: 22957016 PMCID: PMC3434197 DOI: 10.1371/journal.pone.0041559] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Accepted: 06/26/2012] [Indexed: 11/19/2022] Open
Abstract
Because cross-species evidence suggests that high testosterone (T) may interfere with paternal investment, the relationships between men's transition to parenting and changes in their T are of growing interest. Studies of human males suggest that fathers who provide childcare often have lower T than uninvolved fathers, but no studies to date have evaluated how nighttime sleep proximity between fathers and their offspring may affect T. Using data collected in 2005 and 2009 from a sample of men (n = 362; age 26.0 ± 0.3 years in 2009) residing in metropolitan Cebu, Philippines, we evaluated fathers' T based on whether they slept on the same surface as their children (same surface cosleepers), slept on a different surface but in the same room (roomsharers), or slept separately from their children (solitary sleepers). A large majority (92%) of fathers in this sample reported practicing same surface cosleeping. Compared to fathers who slept solitarily, same surface cosleeping fathers had significantly lower evening (PM) T and also showed a greater diurnal decline in T from waking to evening (both p<0.05). Among men who were not fathers at baseline (2005), fathers who were cosleepers at follow-up (2009) experienced a significantly greater longitudinal decline in PM T over the 4.5-year study period (p<0.01) compared to solitary sleeping fathers. Among these same men, baseline T did not predict fathers' sleeping arrangements at follow-up (p>0.2). These results are consistent with previous findings indicating that daytime father-child interaction contributes to lower T among fathers. Our findings specifically suggest that close sleep proximity between fathers and their offspring results in greater longitudinal decreases in T as men transition to fatherhood and lower PM T overall compared to solitary sleeping fathers.
Collapse
Affiliation(s)
- Lee T Gettler
- Department of Anthropology, University of Notre Dame, Notre Dame, Indiana, United States of America.
| | | | | | | | | |
Collapse
|
21
|
Mammi C, Calanchini M, Antelmi A, Cinti F, Rosano GMC, Lenzi A, Caprio M, Fabbri A. Androgens and adipose tissue in males: a complex and reciprocal interplay. Int J Endocrinol 2012; 2012:789653. [PMID: 22235202 PMCID: PMC3253446 DOI: 10.1155/2012/789653] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Accepted: 09/15/2011] [Indexed: 12/28/2022] Open
Abstract
Clinical evidence shows that in males obesity is frequently associated with hypogonadism and vice versa; also, low testosterone levels have been considered a "hallmark" of metabolic syndrome in men. These observations indicate that there is a strict connection between anatomically and functionally distinct cell types such as white adipocytes and Leydig cells, that synthesize testosterone. Adipose tissue is able to control several functions of the testis through its products secreted in the bloodstream. On the other hand, circulating levels of testosterone and estradiol deeply affect adipocyte proliferation, differentiation, and fat mass distribution, hereby controlling critical metabolic functions, such as food intake, insulin sensitivity, vascular reactivity, and immunity. This paper highlights the existing clinical and experimental evidence linking androgens and adipose tissue and illustrates the consequences occurring when the balance between fat mass distribution and eugonadism is lost.
Collapse
Affiliation(s)
- Caterina Mammi
- Centre of Clinical and Basic Research, Department of Medical Sciences, IRCCS San Raffaele Pisana, 235 00163 Rome, Italy
- *Caterina Mammi:
| | - Matilde Calanchini
- Unit of Endocrinology, S. Eugenio & CTO A. Alesini Hospitals, Department of Internal Medicine, University of Rome “Tor Vergata”, Rome, Italy
| | - Antonella Antelmi
- Centre of Clinical and Basic Research, Department of Medical Sciences, IRCCS San Raffaele Pisana, 235 00163 Rome, Italy
| | - Francesca Cinti
- Centre of Clinical and Basic Research, Department of Medical Sciences, IRCCS San Raffaele Pisana, 235 00163 Rome, Italy
| | - Giuseppe M. C. Rosano
- Centre of Clinical and Basic Research, Department of Medical Sciences, IRCCS San Raffaele Pisana, 235 00163 Rome, Italy
| | - Andrea Lenzi
- Department of Medical Pathophysiology, University of Rome “La Sapienza”, Rome, Italy
| | - Massimiliano Caprio
- Centre of Clinical and Basic Research, Department of Medical Sciences, IRCCS San Raffaele Pisana, 235 00163 Rome, Italy
| | - Andrea Fabbri
- Unit of Endocrinology, S. Eugenio & CTO A. Alesini Hospitals, Department of Internal Medicine, University of Rome “Tor Vergata”, Rome, Italy
| |
Collapse
|
22
|
Lee S, Braden B, Kang SS, Rivier C. Urocortins are present in the rat testis. Neuropeptides 2011; 45:131-7. [PMID: 21256589 PMCID: PMC3043384 DOI: 10.1016/j.npep.2010.12.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Revised: 11/11/2010] [Accepted: 12/19/2010] [Indexed: 10/18/2022]
Abstract
The synthesis and release of testosterone (T) depends both on circulating luteinizing hormone (LH) and on an array of testicular factors whose role remains incompletely understood. Corticotropin-releasing factor (CRF) had been reported in the rat testes, where it was thought to inhibit T secretion. However, the discovery that the CRF-related peptides urocortins (Ucns), of which there are currently three subtypes (Ucn 1, 2 and 3), cross-react with many reagents previously used to detect CRF, has cast doubt on this concept. Here we show that while CRF was readily measurable in rat hypothalami (which served as controls), signals for this peptide were barely detectable in total RNA extracted from the testes. On the other hand, microarray, RT-PCR and real-time quantitative RT-PCR (qRT-PCR) analyses all indicated strong signals for Ucn 1 in the male gonads, with weaker levels of Ucn 2 and 3 mRNA gene expression. Results obtained for Ucn 1 gene expression were corroborated by immunohistochemical detection, which appeared restricted to Leydig cells. Finally, to investigate possible changes in testicular Ucn 1 levels induced by homeostatic challenges, we measured them in rats exposed to alcohol. We observed that indeed, the intragastric injection of this drug significantly increased testicular Ucn 1, but not Ucn 2, Ucn 3, CRF, CRFR1 or CRFR2 mRNA levels. Collectively, these results provide novel information regarding the presence of CRF-like peptides in the adult male rat testis.
Collapse
Affiliation(s)
- Soon Lee
- The Clayton Foundation Laboratories for Peptide Biology, The Salk Institute, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Brian Braden
- The Clayton Foundation Laboratories for Peptide Biology, The Salk Institute, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Sang Soo Kang
- Department of Anatomy & Neurobiology, Health Science Institute, School of Medicine, Gyeongsang National University, 92 Chilam-Dong, Jinju, Gyeongnam 660-751, Korea
| | - Catherine Rivier
- The Clayton Foundation Laboratories for Peptide Biology, The Salk Institute, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| |
Collapse
|
23
|
Lagarrigue M, Becker M, Lavigne R, Deininger SO, Walch A, Aubry F, Suckau D, Pineau C. Revisiting rat spermatogenesis with MALDI imaging at 20-microm resolution. Mol Cell Proteomics 2010; 10:M110.005991. [PMID: 21149303 DOI: 10.1074/mcp.m110.005991] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Matrix-assisted laser desorption/ionization (MALDI) molecular imaging technology attracts increasing attention in the field of biomarker discovery. The unambiguous correlation between histopathology and MALDI images is a key feature for success. MALDI imaging mass spectrometry (MS) at high definition thus calls for technological developments that were established by a number of small steps. These included tissue and matrix preparation steps, dedicated lasers for MALDI imaging, an increase of the robustness against cell debris and matrix sublimation, software for precision matching of molecular and microscopic images, and the analysis of MALDI imaging data using multivariate statistical methods. The goal of these developments is to approach single cell resolution with imaging MS. Currently, a performance level of 20-μm image resolution was achieved with an unmodified and commercially available instrument for proteins detected in the 2-16-kDa range. The rat testis was used as a relevant model for validating and optimizing our technological developments. Indeed, testicular anatomy is among the most complex found in mammalian bodies. In the present study, we were able to visualize, at 20-μm image resolution level, different stages of germ cell development in testicular seminiferous tubules; to provide a molecular correlate for its well established stage-specific classification; and to identify proteins of interest using a top-down approach and superimpose molecular and immunohistochemistry images.
Collapse
Affiliation(s)
- Mélanie Lagarrigue
- INSERM U625, Proteomics Core Facility Biogenouest, Campus de Beaulieu, Université de Rennes I, F-35042 Rennes, France
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Gonzalez CR, Matzkin ME, Frungieri MB, Terradas C, Ponzio R, Puigdomenech E, Levalle O, Calandra RS, Gonzalez-Calvar SI. Expression of the TGF-beta1 system in human testicular pathologies. Reprod Biol Endocrinol 2010; 8:148. [PMID: 21126344 PMCID: PMC3009701 DOI: 10.1186/1477-7827-8-148] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2010] [Accepted: 12/02/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In non-obstructive azoospermia, histological patterns of Sertoli cell-only Syndrome (SCO) and hypospermatogenesis (H) are commonly found. In these pathologies, Leydig cell hyperplasia (LCH) is detected in some patients. Since TGF-β1 is involved in cellular proliferation/development, the aim of this work was to analyze the expression of TGF-β1, its receptors TGFBRII, TGFBRI (ALK-1 and ALK-5), and the co-receptor endoglin in human biopsies from patients with idiopathic infertility. METHODS Specific immunostaining of TGF-β1, its receptors TGFBRII, TGFBRI (ALK-1 and ALK-5), co-receptor endoglin and Smads proteins, were carried out in testicular biopsies from normal and infertile men with SCO or H. Gene expression of TGF-β1 system were made in biopsies from infertile patients with semi-quantitative and quantitative PCR. RESULTS Immunohistochemical studies revealed that TGF-β1 and its specific receptors are present in Leydig cells in biopsies from normal tissue or patients with SCO or H with or without LCH. Smad proteins, which are involved in TGF-β1 signaling, are also detected in both their phosphorylated (activated) and dephosphorylated form in all samples TGF-β1, ALK-1 and endoglin gene expression are stronger in human biopsies with LCH than in those with SCO or H. Neither TGFBRII nor ALK-5 gene expression showed significant differences between pathologies. A significant correlation between ALK-1 and endoglin expression was observed. CONCLUSIONS In conclusion, the high levels of mRNA and protein expression of the TGF-β1 system in patients with LCH, particularly ALK1 and its correlation with endoglin, suggest that these proteins acting in concert might be, at least in part, committed actors in the Leydig cell hyperplasia.
Collapse
Affiliation(s)
- Candela R Gonzalez
- Instituto de Biología y Medicina Experimental (CONICET), Vuelta de Obligado 2490 (1428) Buenos Aires, Argentina
| | - María E Matzkin
- Instituto de Biología y Medicina Experimental (CONICET), Vuelta de Obligado 2490 (1428) Buenos Aires, Argentina
- Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155 (1121) Buenos Aires, Argentina
| | - Mónica B Frungieri
- Instituto de Biología y Medicina Experimental (CONICET), Vuelta de Obligado 2490 (1428) Buenos Aires, Argentina
- Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155 (1121) Buenos Aires, Argentina
| | - Claudio Terradas
- División Endocrinología, Hospital Durand, Av. Díaz Velez 5044 (1405) Buenos Aires, Argentina
- Instituto Medico PREFER, Calle 995 N° 2348, San Martín (6000) Argentina
| | - Roberto Ponzio
- Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155 (1121) Buenos Aires, Argentina
| | | | - Oscar Levalle
- División Endocrinología, Hospital Durand, Av. Díaz Velez 5044 (1405) Buenos Aires, Argentina
| | - Ricardo S Calandra
- Instituto de Biología y Medicina Experimental (CONICET), Vuelta de Obligado 2490 (1428) Buenos Aires, Argentina
| | - Silvia I Gonzalez-Calvar
- Instituto de Biología y Medicina Experimental (CONICET), Vuelta de Obligado 2490 (1428) Buenos Aires, Argentina
- Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155 (1121) Buenos Aires, Argentina
| |
Collapse
|
25
|
Collins JJ, Hou X, Romanova EV, Lambrus BG, Miller CM, Saberi A, Sweedler JV, Newmark PA. Genome-wide analyses reveal a role for peptide hormones in planarian germline development. PLoS Biol 2010; 8:e1000509. [PMID: 20967238 PMCID: PMC2953531 DOI: 10.1371/journal.pbio.1000509] [Citation(s) in RCA: 195] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2010] [Accepted: 08/25/2010] [Indexed: 12/02/2022] Open
Abstract
Genomic/peptidomic analyses of the planarian Schmidtea mediterranea identifies >200 neuropeptides and uncovers a conserved neuropeptide required for proper maturation and maintenance of the reproductive system. Bioactive peptides (i.e., neuropeptides or peptide hormones) represent the largest class of cell-cell signaling molecules in metazoans and are potent regulators of neural and physiological function. In vertebrates, peptide hormones play an integral role in endocrine signaling between the brain and the gonads that controls reproductive development, yet few of these molecules have been shown to influence reproductive development in invertebrates. Here, we define a role for peptide hormones in controlling reproductive physiology of the model flatworm, the planarian Schmidtea mediterranea. Based on our observation that defective neuropeptide processing results in defects in reproductive system development, we employed peptidomic and functional genomic approaches to characterize the planarian peptide hormone complement, identifying 51 prohormone genes and validating 142 peptides biochemically. Comprehensive in situ hybridization analyses of prohormone gene expression revealed the unanticipated complexity of the flatworm nervous system and identified a prohormone specifically expressed in the nervous system of sexually reproducing planarians. We show that this member of the neuropeptide Y superfamily is required for the maintenance of mature reproductive organs and differentiated germ cells in the testes. Additionally, comparative analyses of our biochemically validated prohormones with the genomes of the parasitic flatworms Schistosoma mansoni and Schistosoma japonicum identified new schistosome prohormones and validated half of all predicted peptide-encoding genes in these parasites. These studies describe the peptide hormone complement of a flatworm on a genome-wide scale and reveal a previously uncharacterized role for peptide hormones in flatworm reproduction. Furthermore, they suggest new opportunities for using planarians as free-living models for understanding the reproductive biology of flatworm parasites. Flatworms cause diseases affecting hundreds of millions of people, so understanding what influences their reproductive activity is of fundamental importance. Neurally derived signals have been suggested to coordinate sexual reproduction in free-living flatworms, yet the neuroendocrine signaling repertoire has not been characterized comprehensively for any flatworm. Neuropeptides are a large diverse group of cell-cell signaling molecules and play many roles in vertebrate reproductive development; however, little is known about their function in reproductive development among invertebrates. Here we use biochemical and bioinformatic techniques to identify bioactive peptides in the genome of the planarian flatworm Schmidtea mediterranea and identify 51 genes encoding >200 peptides. Analysis of these genes in both sexual and asexual strains of S. mediterranea identified a neuropeptide Y superfamily member as important for the normal development and maintenance of the planarian reproductive system. We suggest that understanding peptide hormone function in planarian reproduction could have practical implications in the treatment of parasitic flatworms.
Collapse
Affiliation(s)
- James J. Collins
- Howard Hughes Medical Institute and Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Xiaowen Hou
- Center for Biophysics and Computational Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Elena V. Romanova
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Bramwell G. Lambrus
- Howard Hughes Medical Institute and Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Claire M. Miller
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Amir Saberi
- Howard Hughes Medical Institute and Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Jonathan V. Sweedler
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Phillip A. Newmark
- Howard Hughes Medical Institute and Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- * E-mail:
| |
Collapse
|
26
|
Rolland AD, Lareyre JJ, Goupil AS, Montfort J, Ricordel MJ, Esquerré D, Hugot K, Houlgatte R, Chalmel F, Le Gac F. Expression profiling of rainbow trout testis development identifies evolutionary conserved genes involved in spermatogenesis. BMC Genomics 2009; 10:546. [PMID: 19925684 PMCID: PMC2786911 DOI: 10.1186/1471-2164-10-546] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2009] [Accepted: 11/20/2009] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Spermatogenesis is a late developmental process that involves a coordinated expression program in germ cells and a permanent communication between the testicular somatic cells and the germ-line. Current knowledge regarding molecular factors driving male germ cell proliferation and differentiation in vertebrates is still limited and mainly based on existing data from rodents and human. Fish with a marked reproductive cycle and a germ cell development in synchronous cysts have proven to be choice models to study precise stages of the spermatogenetic development and the germ cell-somatic cell communication network. In this study we used 9K cDNA microarrays to investigate the expression profiles underlying testis maturation during the male reproductive cycle of the trout, Oncorhynchus mykiss. RESULTS Using total testis samples at various developmental stages and isolated spermatogonia, spermatocytes and spermatids, 3379 differentially expressed trout cDNAs were identified and their gene activation or repression patterns throughout the reproductive cycle were reported. We also performed a tissue-profiling analysis and highlighted many genes for which expression signals were restricted to the testes or gonads from both sexes. The search for orthologous genes in genome-sequenced fish species and the use of their mammalian orthologs allowed us to provide accurate annotations for trout cDNAs. The analysis of the GeneOntology terms therefore validated and broadened our interpretation of expression clusters by highlighting enriched functions that are consistent with known sequential events during male gametogenesis. Furthermore, we compared expression profiles of trout and mouse orthologs and identified a complement of genes for which expression during spermatogenesis was maintained throughout evolution. CONCLUSION A comprehensive study of gene expression and associated functions during testis maturation and germ cell differentiation in the rainbow trout is presented. The study identifies new pathways involved during spermatogonia self-renewal or rapid proliferation, meiosis and gamete differentiation, in fish and potentially in all vertebrates. It also provides the necessary basis to further investigate the hormonal and molecular networks that trigger puberty and annual testicular recrudescence in seasonally breeding species.
Collapse
Affiliation(s)
| | | | | | - Jérôme Montfort
- INRA, UR1037, IFR-140, Ouest Genopole, Rennes, 35042, France
| | | | - Diane Esquerré
- INRA, UMR 1313 de Génétique Animale et Biologie Intégrative, Domaine de Vilvert, 78350 Jouy-en-Josas, France
- CEA, DSV, iRCM, SREIT, Laboratoire de Génétique Animale et Biologie Intégrative, 78350 Jouy-en-Josas, France
- AgroParisTech, UMR de Génétique Animale et Biologie Intégrative, 78350 Jouy-en-Josas, France
| | - Karine Hugot
- INRA, UMR 1313 de Génétique Animale et Biologie Intégrative, Domaine de Vilvert, 78350 Jouy-en-Josas, France
- CEA, DSV, iRCM, SREIT, Laboratoire de Génétique Animale et Biologie Intégrative, 78350 Jouy-en-Josas, France
- AgroParisTech, UMR de Génétique Animale et Biologie Intégrative, 78350 Jouy-en-Josas, France
| | - Rémi Houlgatte
- Inserm, U915, Université de Nantes, Faculté de Médecine, Nantes, 44035, France
| | - Fréderic Chalmel
- Inserm, U625, GERHM, Université de Rennes1, IFR-140, Ouest Genopole, Rennes, 35042, France
| | - Florence Le Gac
- INRA, UR1037, IFR-140, Ouest Genopole, Rennes, 35042, France
| |
Collapse
|
27
|
Schmahl J, Rizzolo K, Soriano P. The PDGF signaling pathway controls multiple steroid-producing lineages. Genes Dev 2009; 22:3255-67. [PMID: 19056881 DOI: 10.1101/gad.1723908] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The platelet-derived growth factor (PDGF) signaling pathway regulates numerous lineages of mesenchymal cell origin during development and in the adult. The transcriptional targets of this pathway have been shown to be required in several PDGF-dependent processes, but the roles of these targets in specific tissues is just beginning to be identified. In this study, we show that five different PDGF target genes are essential for male and/or female fertility. Mutations in each of these five different genes lead to defects in the steroid-producing cells in the testis and/or ovary and altered hormone production, suggesting that the PDGF pathway controls steroidogenesis through these genes in both sexes. Furthermore, conditional mutations of both PDGF receptors revealed a requirement in steroid-producing cells in multiple organs, including the testis, ovary, and adrenal cortex. Therefore, PDGF signaling may constitute a common mechanism in the control of multiple steroidogenic lineages.
Collapse
Affiliation(s)
- Jennifer Schmahl
- Program in Developmental Biology, Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | | | | |
Collapse
|
28
|
Abstract
Corticotropin-releasing factor (CRF) has previously been reported in rat testes in which it inhibits Leydig cells activity. However, recent studies in our laboratory have suggested that some of the effects originally attributed to CRF were instead due to the related peptide Urocortin 1 (Ucn 1) and that this latter hormone, not CRF, was detectable in Leydig cells. We show here that Ucn 1 [a mixed CRF receptor (CRFR) type 1 and CRFR2 agonist] and the CRFR1-selective peptide Stressin 1, but not Ucn 2 or Ucn 3 (both considered selective CRFR2 ligands), significantly blunt the testosterone response to human chorionic gonadotropin. The effect of Ucn 1 is observed regardless of whether this peptide is injected iv or directly into the testes, and it is reversed by the mixed CRFR1/R2 antagonist Astressin B. Blockade of GnRH receptors with the antagonist Azalin B does not interfere with the influence of Ucn 1, thereby demonstrating that pituitary luteinizing hormone does not appear to be involved in this model. Collectively these results suggest that Ucn 1, not CRF, is present in the rat testes and interferes with Leydig cell activity. However, whereas we previously reported that alcohol up-regulated gonadal Ucn 1 gene expression, CRF receptor antagonists were unable to reverse the inhibitory effect exerted by alcohol on human chorionic gonadotropin-induced testosterone release. The functional role played by testicular Ucn 1 in stress models characterized by blunted androgen levels therefore needs to be further investigated.
Collapse
Affiliation(s)
- Catherine L Rivier
- The Clayton Foundation Laboratories for Peptide Biology, The Salk Institute, La Jolla, California 92037, USA.
| |
Collapse
|
29
|
Rezvani A, Ozaras N. Infertility improved by etanercept in ankylosing spondylitis. Indian J Pharmacol 2008; 40:276-7. [PMID: 21279185 PMCID: PMC3025146 DOI: 10.4103/0253-7613.45155] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2007] [Revised: 01/31/2008] [Accepted: 11/22/2008] [Indexed: 11/11/2022] Open
Abstract
The effect of TNF-α and TNF-α antagonists on semen quality in men is controversial. TNF-α levels are usually low in seminal plasma, but they tend to increase in inflammatory and infectious diseases. Etanercept is a highly-specific antagonist of TNF-α. In this report, we describe the development of pregnancy in a couple with a previously infertile husband, who received etanercept for ankylosing spondylitis.
Collapse
Affiliation(s)
- Aylin Rezvani
- Physical Medicine and Rehabilitation Department, Vakif Gureba Training Hospital, Istanbul, Turkey
| | - Nihal Ozaras
- Physical Medicine and Rehabilitation Department, Vakif Gureba Training Hospital, Istanbul, Turkey
| |
Collapse
|
30
|
Usdin TB, Paciga M, Riordan T, Kuo J, Parmelee A, Petukova G, Camerini-Otero RD, Mezey E. Tuberoinfundibular Peptide of 39 residues is required for germ cell development. Endocrinology 2008; 149:4292-300. [PMID: 18483145 PMCID: PMC2553379 DOI: 10.1210/en.2008-0419] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Tuberoinfundibular peptide of 39 residues (TIP39) was identified as a PTH 2 receptor ligand. We report that mice with deletion of Tifp39, the gene encoding TIP39, are sterile. Testes contained Leydig and Sertoli cells and spermatogonia but no spermatids. Labeling chromosome spreads with antibodies to proteins involved in recombination showed that spermatogonia do not complete prophase of meiosis I. Chromosomes were observed at different stages of recombination in single nuclei, a defect not previously described with mutations in genes known to be specifically involved in DNA replication and recombination. TIP39 was previously shown to be expressed in neurons projecting to the hypothalamus and within the testes. LH and FSH were slightly elevated in Tifp39(-/-) mice, suggesting intact hypothalamic function. We found using in situ hybridization that the genes encoding TIP39 and the PTH 2 receptor are expressed in a stage-specific manner within seminiferous tubules. Using immunohistochemistry and quantitative RT-PCR, TIP39 expression is greatest in mature testes, and appears most abundant in postmeiotic spermatids, but TIP39 protein and mRNA can be detected before any cells have completed meiosis. We used mice that express Cre recombinase under control of a spermatid-specific promoter to express selectively a cDNA encoding TIP39 in the testes of Tifp39(-/-) mice. Spermatid production and fertility were rescued, demonstrating that the defect in Tifp39(-/-) mice was due to the loss of TIP39. These results show that TIP39 is essential for germ cell development and suggest that it may act as an autocrine or paracrine agent within the gonads.
Collapse
Affiliation(s)
- Ted B Usdin
- National Institute of Mental Health/National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Purdue MP, Sakoda LC, Graubard BI, Welch R, Chanock SJ, Sesterhenn IA, Rubertone MV, Erickson RL, McGlynn KA. A case-control investigation of immune function gene polymorphisms and risk of testicular germ cell tumors. Cancer Epidemiol Biomarkers Prev 2007; 16:77-83. [PMID: 17220333 DOI: 10.1158/1055-9965.epi-06-0573] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
There is reason to suspect that testicular germ cell tumor (TGCT) development may be influenced by cytokines, secreted proteins that modulate tumor immune surveillance activity as well as a variety of processes in the testis. To address this hypothesis, we conducted a case-control analysis (508 cases, 608 controls) of 32 putatively functional single-nucleotide polymorphisms (SNP) in 16 immune function genes among non-Hispanic Caucasian participants in the U.S. Servicemen's Testicular Tumor Environmental and Endocrine Determinants Study. The TGFB1 Ex5-73C>T variant was positively associated with TGCT (CT/TT versus CC: odds ratio, 1.73; 95% confidence interval, 1.01-2.95; P(trend) = 0.05); additionally, haplotypes of the assessed TGFB1 SNPs (-509C>T, 327C>T, Ex1-282C>G, and Ex5-73C>T) differed in frequency between cases and controls (all TGCT, P 0.07; seminoma, P 0.04; nonseminoma, P 0.11). We also observed excess frequencies among TGCT cases versus controls of LTA 252G (P(trend) = 0.08) and of the TNF variants -1042C (P(trend) = 0.06), -1036T (P(trend) = 0.07), and -238G (P(trend) = 0.09). Analyses of haplotypes for LTA-TNF SNPs (LTA -91C>A, LTA 252A>G, TNF -863C>A, TNF -857C>T, TNF -308G>A, and -238G>A) were similarly suggestive of an association with TGCT (P = 0.06) and nonseminoma (P = 0.04), but not seminoma (P = 0.21). Polymorphisms in other genes were found to be associated only with seminoma (IL2) or nonseminoma (IFNGR2 and IL10). However, none of the associations remained noteworthy after applying the false discovery rate method to control for multiple testing. In conclusion, our findings suggest that polymorphisms in TGFB1 and LTA/TNF, and possibly other immune function genes, may influence susceptibility to TGCT.
Collapse
Affiliation(s)
- Mark P Purdue
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Department of Health and Human Services, EPS-8009, 6120 Executive Boulevard, Rockville, MD 20892-7234, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Behr R, Sackett SD, Bochkis IM, Le PP, Kaestner KH. Impaired male fertility and atrophy of seminiferous tubules caused by haploinsufficiency for Foxa3. Dev Biol 2007; 306:636-45. [PMID: 17488644 PMCID: PMC1952241 DOI: 10.1016/j.ydbio.2007.03.525] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2007] [Revised: 03/15/2007] [Accepted: 03/30/2007] [Indexed: 11/22/2022]
Abstract
Foxa1, 2 and 3 (formerly HNF-3alpha, -beta and -gamma) constitute a sub-family of winged helix transcription factors with multiple roles in mammalian organ development. While all three Foxa mRNAs are present in endoderm derivatives including liver and pancreas, only Foxa3 is expressed in the testis. Here we demonstrate by genetic lineage tracing that Foxa3 is expressed in postmeiotic germ and interstitial Leydig cells. The germinal epithelium of Foxa3-deficient testes is characterized by a loss of germ cells secondary to an increase in germ cell apoptosis that ultimately leads to a Sertoli cell-only syndrome. Remarkably, not only the Foxa3(-/-) mice but also Foxa3(+/-) mice exhibited loss of germ cells. This cellular phenotype caused significantly reduced fertility and testis weight of both Foxa3(-/-) and Foxa3(+/-) mice. Using microarray analysis, we found a dramatic downregulation of the zinc finger protein 93 and the testicular tumor-associated paraneoplastic Ma antigen (PNMA) and increased expression of a number of genes including zinc finger protein 94 and several kallikrein 1-related peptidases which could account for at least part of the observed phenotype. In summary, we have identified Foxa3 as a transcriptional regulator with a dominant phenotype in germ cell maintenance and suggest FOXA3 as a potential candidate gene for subfertility in man.
Collapse
Affiliation(s)
- Rüdiger Behr
- University of Pennsylvania Medical School, Department of Genetics, 560 CRB, 415 Curie Blvd., Philadelphia, PA 19104 USA
| | - Sara D. Sackett
- University of Pennsylvania Medical School, Department of Genetics, 560 CRB, 415 Curie Blvd., Philadelphia, PA 19104 USA
| | - Irina M. Bochkis
- University of Pennsylvania Medical School, Department of Genetics, 560 CRB, 415 Curie Blvd., Philadelphia, PA 19104 USA
| | - Phillip Phuc Le
- University of Pennsylvania Medical School, Department of Genetics, 560 CRB, 415 Curie Blvd., Philadelphia, PA 19104 USA
| | - Klaus H. Kaestner
- University of Pennsylvania Medical School, Department of Genetics, 560 CRB, 415 Curie Blvd., Philadelphia, PA 19104 USA
| |
Collapse
|
33
|
Kassab M, Abd-Elmaksoud A, Ali MA. Localization of the epidermal growth factor (EGF) and epidermal growth factor receptor (EGFR) in the bovine testis. J Mol Histol 2007; 38:207-14. [PMID: 17492480 DOI: 10.1007/s10735-007-9089-2] [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] [Received: 02/27/2007] [Accepted: 03/22/2007] [Indexed: 10/23/2022]
Abstract
In the last few decades, several growth factors were identified in the testis of various mammalian species. Growth factors are shown to promote cell proliferation, regulate tissue differentiation, and modulate organogenesis. In the present investigation we have studied the localization of EGF and EGFR in the adult bovine testis by means of immunohistochemical method. Our results demonstrated that EGF and EGFR were localized solely to the bovine testicular germ cells (spermatogonia, spermatocytes, and round spermatids). In contrast, the somatic testicular cells (i.e., Sertoli, Leydig, and myofibroblast cells) exhibited no staining affinity. EGF and EGFR were additionally detected in the epithelial lining of straight tubules and rete testis. Interestingly, the distribution of EGF and EGFR in the germ cells was mainly dependent upon the cycle of the seminiferous epithelium since their localization appeared to be preponderant during the spermatogonia proliferation and during the meiotic and spermiogenic processes. In conclusion, such findings may suggest that EGF and EGFR are important paracrine and/or autocrine regulators of spermatogenesis in bovine.
Collapse
Affiliation(s)
- M Kassab
- Department of Anatomy and Histology, Faculty of Veterinary Medicine, Kafr El-Sheikh University, Kafr El-Sheikh, Egypt
| | | | | |
Collapse
|
34
|
Xia W, Mruk DD, Cheng CY. C-type natriuretic peptide regulates blood-testis barrier dynamics in adult rat testes. Proc Natl Acad Sci U S A 2007; 104:3841-6. [PMID: 17360440 PMCID: PMC1820671 DOI: 10.1073/pnas.0610100104] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2006] [Indexed: 12/13/2022] Open
Abstract
In adult rat testes, the blood-testis barrier (BTB) in the seminiferous epithelium must "open" (or "disassemble") to accommodate the migration of preleptotene spermatocytes from the basal to the adluminal compartment that occurs at stage VIII of the epithelial cycle. However, the molecule(s) and/or mechanism(s) that regulate this event are unknown. In this report, C-type natriuretic peptide (CNP) was shown to be a regulator of BTB dynamics. Although Sertoli and germ cells contributed to the pool of CNP in the seminiferous epithelium, its receptor, natriuretic peptide receptor B, resided almost exclusively in Sertoli cells. CNP also expressed stage-specifically and localized predominantly at the BTB in the seminiferous epithelium at stage VIII of the epithelial cycle. A synthetic CNP-22 peptide, when added to Sertoli cell cultures, was shown to perturb Sertoli cell tight junction in vitro, causing disappearance of BTB-associated proteins (JAM-A, occludin, N-cadherin, and beta-catenin) from the cell-cell interface. This inhibitory effect of CNP on the tight junction was confirmed by transient overexpression of CNP in these cells, which was mediated, at least in part, by accelerating the internalization of BTB integral membrane proteins. To validate these in vitro findings, CNP-22 was administered to testes at a dose of 0.35 or 3.5 mug per testis, which was shown to perturb the BTB integrity In vivo when the barrier function was assessed by monitoring the diffusion of a small molecular probe across the BTB. In summary, CNP secreted by Sertoli and germ cells into the BTB microenvironment regulates BTB dynamics during spermatogenesis.
Collapse
Affiliation(s)
- Weiliang Xia
- Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY 10021
| | - Dolores D. Mruk
- Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY 10021
| | - C. Yan Cheng
- Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY 10021
| |
Collapse
|
35
|
Xia W, Mruk DD, Lee WM, Cheng CY. Unraveling the molecular targets pertinent to junction restructuring events during spermatogenesis using the Adjudin-induced germ cell depletion model. J Endocrinol 2007; 192:563-83. [PMID: 17332525 PMCID: PMC2804028 DOI: 10.1677/joe-06-0158] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
During spermatogenesis, extensive restructuring takes place at the Sertoli-Sertoli and Sertoli-germ cell interface, which is regulated via intriguing interactions among cytokines, proteases, protease inhibitors, kinases, phosphatases, and transcription factors. This in turn determines the steady-state levels of integral membrane proteins at the cell junctions. We sought to further expand these observations using the Adjudin model. Adjudin is a potential male contraceptive that targets Sertoli-germ cell adhesion, causing exfoliation of spermatids and spermatocytes, but not spermatogonia, from the seminiferous epithelium. This model thus provides the means to identify crucial regulatory molecules and signaling pathways pertinent to junction restructuring events during spermatogenesis. In this study, genome-wide expression profiling of rat testes after treatment with Adjudin at the time of extensive junction restructuring was performed. Differentially regulated genes, such as cytokines, proteases, protease inhibitors, cell junction-associated proteins, and transcription factors pertinent to junction restructuring were identified. These data were consistent with earlier findings; however, much new information was obtained which has been deposited at the Gene Expression Omnibus data repository website: http://www.ncbi.nih.gov/geo/ with Accession number: GSE5131. The primary signaling events pertinent to junction restructuring in the testis induced by Adjudin were also delineated using bioinformatics. These findings were also consistent with recently published reports. The identified molecular signatures or targets pertinent to junction dynamics in the testis as reported herein, many of which have not been investigated, thus offer a framework upon which the regulation of junction restructuring events at the Sertoli-Sertoli and Sertoli-germ cell interface pertinent to spermatogenesis can be further studied.
Collapse
Affiliation(s)
- Weiliang Xia
- Center for Biomedical Research, Population Council, 1230 York Avenue, New York, New York 10021, USA
| | | | | | | |
Collapse
|
36
|
Yoshimura Y. Insulin-like growth factors and their binding proteins: Potential relevance to reproductive physiology. Reprod Med Biol 2003; 2:1-24. [PMID: 29699162 DOI: 10.1046/j.1445-5781.2003.00016.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cyclic ovarian follicular development is a complex process that involves proliferation, differentiation, and death of follicle cells. Gonadotropins produced by the pituitary gland have a central role in the regulation of these processes. In addition, a wide range of paracrine and autocrine factors produced in the reproductive organs have been proposed as regulators of reproductive functions. Components of the insulin-like growth factors (IGF) system are widely expressed in the female reproductive tract. The IGFs and their binding proteins play a significant role in several processes of reproductive physiology, including ovarian follicular development, oogenesis and oocyte maturation, ovulation, luteal function, follicular atresia, and testicular function. The majority of these physiological actions of the IGFs are believed to occur via activation of the IGF-I receptor, although the IGF-I effects are modulated by IGF binding proteins (IGFBPs). As much of the data obtained to date have been in the rodent reproductive organs, it may not be possible to directly extrapolate the results to the primate organs. There is a distinct species-difference in the gene expression and functional roles of the IGF-IGFBP system in reproductive organs. However, the disturbance of the IGF-IGFBP system in human reproductive physiology may lead to anovulation, disorders of androgen excess, infertility associated with implantation failure, and male infertility. Further research is needed in domestic animals to determine if manipulation of the IGF-IGFBP system may result in improved reproductive efficiency. As our understanding of the IGF-IGFBP system increases, the uses of human recombinant IGF peptides and IGFBPs as clinical therapy for disease states is becoming a reality. (Reprod Med Biol 2003; 2: 1-24).
Collapse
Affiliation(s)
- Yasunori Yoshimura
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
37
|
Narula A, Kilen S, Ma E, Kroeger J, Goldberg E, Woodruff TK. Smad4 overexpression causes germ cell ablation and leydig cell hyperplasia in transgenic mice. THE AMERICAN JOURNAL OF PATHOLOGY 2002; 161:1723-34. [PMID: 12414519 PMCID: PMC1850786 DOI: 10.1016/s0002-9440(10)64449-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Members of the transforming growth factor-beta (TGF-beta) superfamily play a variety of important roles in testicular development and function. The tumor suppressor gene, Smad4, is a common mediator of TGF-beta, activin, and bone morphogenetic protein-mediated signaling pathways. To investigate the role of the Smad4 gene during testicular development and function, transgenic mice were generated using a Flag-tagged Smad4 gene driven by 180-bp fragment of the Mullerian inhibiting substance upstream promoter sequence. Three Smad4 transgenic founders (A, B, and G) were detected by Southern blot analysis; line B showed the highest expression of the Smad4 transgene and was further studied. The fertility in F1 generation (B) and F2 generation (BB) of the Smad4 transgenic mice was not impaired. However, in the F3 generation (B2x) all animals were impacted by the overexpression of the Smad4 transgene and two kinds of phenotypes were observed. In one group animals were completely infertile, while in the other group animals were fertile and sired the normal number of pups/litter. These groups are designated as infertile and fertile in the text. Histological evaluation of the testes from the infertile group showed variable degrees of Leydig cell hyperplasia, apoptosis of germ cells, spermatogenic arrest, seminiferous tubule degeneration, and infertility. In the fertile group, there was no apparent change in the histology of the testis except for a slight increase in the number of Leydig cells. Serum follicle-stimulating hormone levels in the adult animals of both groups of Smad4 transgenic male mice were not significantly different from normal littermates; however, testosterone levels in both groups were significantly (P < 0.05) increased. These results suggest that overexpression of Smad4 leads to testicular abnormalities and infertility supporting the hypothesis that the TGF-beta signaling pathways are carefully orchestrated during testicular development. In the absence of normal levels of Smad4 testicular function is compromised.
Collapse
Affiliation(s)
- Anita Narula
- Department of Biochemistry, Northwestern University, Evanston, Illinois 60208, USA
| | | | | | | | | | | |
Collapse
|
38
|
Abstract
Inflammatory disease has been established to affect male reproductive function and fertility. Relevant inflammatory diseases include general and chronic infectious diseases as well as localized acute or chronic infections of the male genitourinary tract. Male accessory gland infections account for almost 15% of all cases of male infertility seen in infertility clinics while fertility usually is not a clinical objective among patients with acute systemic infections such as Gram-negative sepsis. Infections of the male accessory glands frequently are associated with increased counts of white blood cells in semen and elevated levels of proinflammatory cytokines in semen and the testis. There is a mounting body of evidence that demonstrates the importance of cytokines and chemokines in the regulation of testicular and glandular function during pathophysiological states as well as under normal physiological conditions when cytokines act as growth and differentiation factors. The purpose of this review is to examine the role of cytokines in the regulation of steroidogenesis and spermatogenesis in the testis under physiological and pathophysiological conditions and considers clinical investigations that help to improve the evaluation and treatment of male infertility.
Collapse
Affiliation(s)
- D B Hales
- Department of Physiology and Biophysics, University of Illinois at Chicago, 60612-7342, USA.
| | | | | |
Collapse
|