1
|
Neto FTL, Flannigan R, Goldstein M. Regulation of Human Spermatogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1288:255-286. [PMID: 34453741 DOI: 10.1007/978-3-030-77779-1_13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Human spermatogenesis (HS) is an intricate network of sequential processes responsible for the production of the male gamete, the spermatozoon. These processes take place in the seminiferous tubules (ST) of the testis, which are small tubular structures considered the functional units of the testes. Each human testicle contains approximately 600-1200 STs [1], and are capable of producing up to 275 million spermatozoa per day [2].
Collapse
Affiliation(s)
| | - Ryan Flannigan
- Department of Urology, Weill Cornell Medicine, New York, NY, USA.,University of British Columbia, Vancouver, BC, Canada
| | - Marc Goldstein
- Department of Urology, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
2
|
Panza S, Giordano F, De Rose D, Panno ML, De Amicis F, Santoro M, Malivindi R, Rago V, Aquila S. FSH-R Human Early Male Genital Tract, Testicular Tumors and Sperm: Its Involvement in Testicular Disorders. Life (Basel) 2020; 10:life10120336. [PMID: 33317204 PMCID: PMC7764367 DOI: 10.3390/life10120336] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/07/2020] [Accepted: 12/08/2020] [Indexed: 12/26/2022] Open
Abstract
The follicle-stimulating hormone receptor (FSH-R) expression was always considered human gonad-specific. The receptor has also been newly detected in extragonadal tissues. In this finding, we evaluated FSH-R expression in the human male early genital tract, in testicular tumors, and in sperm from healthy and varicocele patients. In sperm, we also studied the mechanism of FSH-R action. Immunohystochemistry and Western blot analysis showed FSH-R presence in the first pathways of the human genital tract, in embryonal carcinoma, and in sperm, but it was absent in seminoma and in lower varicocele. In sperm, FSH/FSH-R activity is mediated by G proteins activating the PKA pathway, as we observed by using the H89. It emerged that increasing FSH treatments induced motility, survival, capacitation, and acrosome reaction in both sperm samples. The different FSH-R expression in tumor testicular tissues may be discriminate by tumor histological type. In spermatozoa, FSH-R indicates a direct action of FSH in these cells, which could be beneficial during semen preparation for in vitro fertilization procedures. For instance, FSH positive effects could be relevant in idiopathic infertility and in the clinic surgery of varicocele. In conclusion, FSH-R expression may be considered a molecular marker of testicular disorders.
Collapse
Affiliation(s)
- Salvatore Panza
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria—Arcavacata di Rende, 87036 Cosenza, Italy; (S.P.); (F.G.); (D.D.R.); (M.L.P.); (F.D.A.); (M.S.); (R.M.); (S.A.)
- Centro Sanitario, University of Calabria—Arcavacata di Rende, 87036 Cosenza, Italy
| | - Francesca Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria—Arcavacata di Rende, 87036 Cosenza, Italy; (S.P.); (F.G.); (D.D.R.); (M.L.P.); (F.D.A.); (M.S.); (R.M.); (S.A.)
| | - Daniela De Rose
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria—Arcavacata di Rende, 87036 Cosenza, Italy; (S.P.); (F.G.); (D.D.R.); (M.L.P.); (F.D.A.); (M.S.); (R.M.); (S.A.)
- Centro Sanitario, University of Calabria—Arcavacata di Rende, 87036 Cosenza, Italy
| | - Maria Luisa Panno
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria—Arcavacata di Rende, 87036 Cosenza, Italy; (S.P.); (F.G.); (D.D.R.); (M.L.P.); (F.D.A.); (M.S.); (R.M.); (S.A.)
| | - Francesca De Amicis
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria—Arcavacata di Rende, 87036 Cosenza, Italy; (S.P.); (F.G.); (D.D.R.); (M.L.P.); (F.D.A.); (M.S.); (R.M.); (S.A.)
- Centro Sanitario, University of Calabria—Arcavacata di Rende, 87036 Cosenza, Italy
| | - Marta Santoro
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria—Arcavacata di Rende, 87036 Cosenza, Italy; (S.P.); (F.G.); (D.D.R.); (M.L.P.); (F.D.A.); (M.S.); (R.M.); (S.A.)
- Centro Sanitario, University of Calabria—Arcavacata di Rende, 87036 Cosenza, Italy
| | - Rocco Malivindi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria—Arcavacata di Rende, 87036 Cosenza, Italy; (S.P.); (F.G.); (D.D.R.); (M.L.P.); (F.D.A.); (M.S.); (R.M.); (S.A.)
| | - Vittoria Rago
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria—Arcavacata di Rende, 87036 Cosenza, Italy; (S.P.); (F.G.); (D.D.R.); (M.L.P.); (F.D.A.); (M.S.); (R.M.); (S.A.)
- Correspondence: ; Tel.: +39-09-8449-6210; Fax: +39-09-8449-3271
| | - Saveria Aquila
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria—Arcavacata di Rende, 87036 Cosenza, Italy; (S.P.); (F.G.); (D.D.R.); (M.L.P.); (F.D.A.); (M.S.); (R.M.); (S.A.)
- Centro Sanitario, University of Calabria—Arcavacata di Rende, 87036 Cosenza, Italy
| |
Collapse
|
3
|
Hau RK, Miller SR, Wright SH, Cherrington NJ. Generation of a hTERT-Immortalized Human Sertoli Cell Model to Study Transporter Dynamics at the Blood-Testis Barrier. Pharmaceutics 2020; 12:pharmaceutics12111005. [PMID: 33105674 PMCID: PMC7690448 DOI: 10.3390/pharmaceutics12111005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/19/2020] [Accepted: 10/20/2020] [Indexed: 12/13/2022] Open
Abstract
The blood-testis barrier (BTB) formed by adjacent Sertoli cells (SCs) limits the entry of many chemicals into seminiferous tubules. Differences in rodent and human substrate-transporter selectivity or kinetics can misrepresent conclusions drawn using rodent in vitro models. Therefore, human in vitro models are preferable when studying transporter dynamics at the BTB. This study describes a hTERT-immortalized human SC line (hT-SerC) with significantly increased replication capacity and minor phenotypic alterations compared to primary human SCs. Notably, hT-SerCs retained similar morphology and minimal changes to mRNA expression of several common SC genes, including AR and FSHR. The mRNA expression of most xenobiotic transporters was within the 2-fold difference threshold in RT-qPCR analysis with some exceptions (OAT3, OCT3, OCTN1, OATP3A1, OATP4A1, ENT1, and ENT2). Functional analysis of the equilibrative nucleoside transporters (ENTs) revealed that primary human SCs and hT-SerCs predominantly express ENT1 with minimal ENT2 expression at the plasma membrane. ENT1-mediated uptake of [3H] uridine was linear over 10 min and inhibited by NBMPR with an IC50 value of 1.35 ± 0.37 nM. These results demonstrate that hT-SerCs can functionally model elements of transport across the human BTB, potentially leading to identification of other transport pathways for xenobiotics, and will guide drug discovery efforts in developing effective BTB-permeable compounds.
Collapse
Affiliation(s)
- Raymond K. Hau
- College of Pharmacy, Department of Pharmacology & Toxicology, University of Arizona, Tucson, AZ 85719, USA; (R.K.H.); (S.R.M.)
| | - Siennah R. Miller
- College of Pharmacy, Department of Pharmacology & Toxicology, University of Arizona, Tucson, AZ 85719, USA; (R.K.H.); (S.R.M.)
| | - Stephen H. Wright
- College of Medicine, Department of Physiology, University of Arizona, Tucson, AZ 85719, USA
- Correspondence: (S.H.W.); (N.J.C.); Tel.: +1-(520)-626-4253 (S.H.W.); +1-(520)-626-0219 (N.J.C.)
| | - Nathan J. Cherrington
- College of Pharmacy, Department of Pharmacology & Toxicology, University of Arizona, Tucson, AZ 85719, USA; (R.K.H.); (S.R.M.)
- Correspondence: (S.H.W.); (N.J.C.); Tel.: +1-(520)-626-4253 (S.H.W.); +1-(520)-626-0219 (N.J.C.)
| |
Collapse
|
4
|
Effects of gonadotropins on testis cell subpopulations of newly hatched chicks treated during embryonic development. Anim Reprod Sci 2020; 222:106600. [PMID: 32980652 DOI: 10.1016/j.anireprosci.2020.106600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 09/08/2020] [Accepted: 09/09/2020] [Indexed: 11/24/2022]
Abstract
The aim of the present study was to evaluate histological and stereological changes, as well as the variations in the number and size of cells from diverse cell subpopulations in testes of newly hatched chicks treated with follicle stimulating hormone (FSH) and luteinizing hormone (LH) during embryonic development. Stereological results indicated that in FSH-treated chicks total volume of the tubular compartment constitutes most of the testis. In contrast, the total volume of interstitial tissue constitutes most of the testis of LH-treated chicks. Results indicate that the number of germ and Sertoli cells increases as a result of FSH and LH treatment, but in FSH-treated testis, Sertoli cells were the most numerous cell type in seminiferous tubules; whereas germ cells were the most numerous cell type in testis of LH-treated chicks. Results also indicate there was a larger total volume of Leydig cells in the testes of FSH- and LH-treated chicks. The larger volume of Leydig cells in FSH-treated chicks is due to a larger cellular volume of these cells, and not due to the number, which remains constant. In contrast, in testes of LH-treated chicks, there is a larger number and volume of Leydig cells. These results indicate the testes of chick embryos respond to FSH and LH treatment, with there being modifications in the seminiferous tubules and interstitial tissue, but these changes differ markedly, indicating that FSH and LH have differential effects on chick testes.
Collapse
|
5
|
Pieri NCG, Mançanares ACF, de Souza AF, Fernandes H, Diaza AMG, Bressan FF, Roballo KCS, Casals JB, Binelli M, Ambrósio CE, Dos Santos Martins D. Xenotransplantation of canine spermatogonial stem cells (cSSCs) regulated by FSH promotes spermatogenesis in infertile mice. Stem Cell Res Ther 2019; 10:135. [PMID: 31109365 PMCID: PMC6528206 DOI: 10.1186/s13287-019-1250-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 04/27/2019] [Accepted: 04/30/2019] [Indexed: 01/15/2023] Open
Abstract
Background Xenotransplantation of spermatogonial stem cells (SSCs) has become a popular topic in various research fields because manipulating these cells can provide insights into the mechanisms associated with germ cell lines and the entire spermatogenesis process; moreover, these cells can be used in several biotechnology applications. To achieve successful xenotransplantation, the in vitro microenvironment in which SSCs are cultured should be an ideal microenvironment for self-renewal and similar to the in vivo testicular microenvironment. The age of the donor, the correct spermatogenesis cycle, and the quality of the donor tissue are also important. Although cell culture-related factors, such as the in vitro supplementation of hormonal factors, are known to promote successful xenotransplantation in mice, little is known about the influence of these factors on SSCs in vitro or in vivo in other mammalian species, such as dogs (Canis lupus familiaris). In this context, the goals of this study were to test the effect of follicle-stimulating hormone (FSH) on canine spermatogonial stem cell (cSSC) cultures since this hormone is related to the glial cell-derived neurotrophic factor (GDNF) signaling pathway, which is responsible for the self-renewal and maintenance of these cells in vivo, and to investigate the microenvironment of the SSC culture after FSH supplementation. Additionally, in vivo analyses of transplanted FSH-supplemented cSSCs in the testes of infertile mice were performed to assess the capacity of cSSCs to develop, maintain, and restore spermatogenesis. Methods SSCs from canine prepubertal testes (aged 3 months) were cultured in vitro in the presence of FSH (10 IU L−1). GFRA1 transcript expression was detected to confirm the spermatogonia population in culture and the effect of FSH on these cells. The protein and transcript levels of late germ cell markers (GFRA1, DAZL, STRA8, PLZF, and CD49f) and a pluripotency marker (OCT4) were detected at 72 and 120 h to confirm the cSSC phenotype. In vivo experiments were performed by transplanting GFP+ cSSCs into infertile mice, and a 10-week follow-up was performed. Histological and immunofluorescence analyses were performed to confirm the repopulation capacity after cSSC xenotransplantation in the testis. Results Supplementation with FSH in cell culture increased the number of cSSCs positive for GFRA1. The cSSCs were also positive for the pluripotency and early germline marker OCT4 and the late germline markers PLZF, DAZL, C-kit, and GFRA-1. The in vivo experiments showed that the cSSCs xenotransplanted into infertile mouse testes were able to repopulate germline cells in the seminiferous tubules of mice. Conclusions In conclusion, our results showed for the first time that the treatment of cSSC cultures with FSH can promote in vitro self-renewal, increase the population of germline cells, and possibly influence the success of spermatogenesis in infertile mice in vivo.
Collapse
Affiliation(s)
- Naira Caroline Godoy Pieri
- Department of Surgery, Faculty of Veterinary Medicine and Animal Sciences, University of São Paulo, Av. Prof. Dr. Orlando Marques de Paiva, 87, Sao Paulo, SP, Brazil. .,Department of Reproduction, Faculty of Veterinary Medicine and Animal Sciences, University of São Paulo, Av. Prof. Dr. Orlando Marques de Paiva, 87, Sao Paulo, SP, Brazil.
| | | | - Aline Fernanda de Souza
- Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo, Av. Duque de Caxias Norte, 225, Pirassununga, SP, Brazil
| | - Hugo Fernandes
- Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo, Av. Duque de Caxias Norte, 225, Pirassununga, SP, Brazil
| | - Angela Maria Gonella Diaza
- North Florida Research and Education Center, Institute of Food and Agricultural Sciences, University of Florida, Marianna, FL, USA
| | - Fabiana Fernandes Bressan
- Department of Surgery, Faculty of Veterinary Medicine and Animal Sciences, University of São Paulo, Av. Prof. Dr. Orlando Marques de Paiva, 87, Sao Paulo, SP, Brazil.,Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo, Av. Duque de Caxias Norte, 225, Pirassununga, SP, Brazil
| | - Kelly Cristine Santos Roballo
- Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo, Av. Duque de Caxias Norte, 225, Pirassununga, SP, Brazil.,School of Pharmacy at University of Wyoming, 1000 E. University Avenue, Laramie, 82071, USA
| | - Juliana Barbosa Casals
- Department of Surgery, Faculty of Veterinary Medicine and Animal Sciences, University of São Paulo, Av. Prof. Dr. Orlando Marques de Paiva, 87, Sao Paulo, SP, Brazil
| | - Mario Binelli
- Department of Animal Sciences, University of Florida, L.E. "Red" Larson Building, Bldg. 499, Room 122 C, Gainesville, FL, 32611-0910, USA
| | - Carlos Eduardo Ambrósio
- Department of Surgery, Faculty of Veterinary Medicine and Animal Sciences, University of São Paulo, Av. Prof. Dr. Orlando Marques de Paiva, 87, Sao Paulo, SP, Brazil.,Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo, Av. Duque de Caxias Norte, 225, Pirassununga, SP, Brazil
| | - Daniele Dos Santos Martins
- Department of Surgery, Faculty of Veterinary Medicine and Animal Sciences, University of São Paulo, Av. Prof. Dr. Orlando Marques de Paiva, 87, Sao Paulo, SP, Brazil.,Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo, Av. Duque de Caxias Norte, 225, Pirassununga, SP, Brazil
| |
Collapse
|
6
|
Shlush E, Maghen L, Swanson S, Kenigsberg S, Moskovtsev S, Barretto T, Gauthier-Fisher A, Librach CL. In vitro generation of Sertoli-like and haploid spermatid-like cells from human umbilical cord perivascular cells. Stem Cell Res Ther 2017; 8:37. [PMID: 28202061 PMCID: PMC5312448 DOI: 10.1186/s13287-017-0491-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 01/24/2017] [Accepted: 01/27/2017] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND First trimester (FTM) and term human umbilical cord-derived perivascular cells (HUCPVCs), which are rich sources of mesenchymal stem cells (MSCs), can give rise to Sertoli cell (SC)-like as well as haploid germ cell (GC)-like cells in vitro using culture conditions that recapitulate the testicular niche. Gamete-like cells have been produced ex vivo using pluripotent stem cells as well as MSCs. However, the production of functional gametes from human stem cells has yet to be achieved. METHODS Three independent lines of FTM and term HUCPVCs were cultured using a novel 5-week step-wise in vitro differentiation protocol recapitulating key physiological signals involved in testicular development. SC- and GC-associated phenotypical properties were assessed by real-time polymerase chain reaction (RT-PCR), quantitative PCR immunocytochemistry, flow cytometry, and fluorescence in-situ hybridization (FISH). Functional spermatogonial stem cell-like properties were assessed using a xenotranplantation assay. RESULTS Within 3 weeks of differentiation, two morphologically distinct cell types emerged including large adherent cells and semi-attached round cells. Both early GC-associated markers (VASA, DAZL, GPR125, GFR1α) and SC-associated markers (FSHR, SOX9, AMH) were upregulated, and 5.7 ± 1.2% of these cells engrafted near the inner basal membrane in a xenograft assay. After 5 weeks in culture, 10-30% of the cells were haploid, had adopted a spermatid-like morphology, and expressed PRM1, Acrosin, and ODF2. Undifferentiated HUCPVCs secreted key factors known to regulate spermatogenesis (LIF, GDNF, BMP4, bFGF) and 10-20% of HUCPVCs co-expressed SSEA4, CD9, CD90, and CD49f. We hypothesize that the paracrine properties and cellular heterogeneity of HUCPVCs may explain their dual capacity to differentiate to both SC- and GC-like cells. CONCLUSIONS HUCPVCs recapitulate elements of the testicular niche including their ability to differentiate into cells with Sertoli-like and haploid spermatid-like properties in vitro. Our study supports the importance of generating a niche-like environment under ex vivo conditions aiming at creating mature GC, and highlights the plasticity of HUCPVCs. This could have future applications for the treatment of some cases of male infertility.
Collapse
Affiliation(s)
- Ekaterina Shlush
- CReATe Fertility Centre, 790 Bay Street, Toronto, Ontario, M5N 1G8, Canada.
| | - Leila Maghen
- CReATe Fertility Centre, 790 Bay Street, Toronto, Ontario, M5N 1G8, Canada
| | - Sonja Swanson
- CReATe Fertility Centre, 790 Bay Street, Toronto, Ontario, M5N 1G8, Canada
| | - Shlomit Kenigsberg
- CReATe Fertility Centre, 790 Bay Street, Toronto, Ontario, M5N 1G8, Canada
| | - Sergey Moskovtsev
- CReATe Fertility Centre, 790 Bay Street, Toronto, Ontario, M5N 1G8, Canada.,Department of Obstetrics & Gynaecology, University of Toronto, Toronto, Ontario, Canada
| | - Tanya Barretto
- CReATe Fertility Centre, 790 Bay Street, Toronto, Ontario, M5N 1G8, Canada
| | | | - Clifford L Librach
- CReATe Fertility Centre, 790 Bay Street, Toronto, Ontario, M5N 1G8, Canada. .,Department of Obstetrics & Gynaecology, University of Toronto, Toronto, Ontario, Canada. .,Department of Physiology, University of Toronto, Toronto, Ontario, Canada. .,Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada. .,Department of Gynecology, Women's College Hospital, Toronto, Ontario, Canada.
| |
Collapse
|
7
|
Herrera-Luna CV, Scarlet D, Walter I, Aurich C. Effect of stallion age on the expression of LH and FSH receptors and aromatase P450 in equine male reproductive tissues. Reprod Fertil Dev 2016; 28:2016-2026. [DOI: 10.1071/rd15027] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 06/06/2015] [Indexed: 11/23/2022] Open
Abstract
The aim of the present study was to characterise receptors for LH and FSH (LHR and FSHR, respectively) and aromatase in epididymal and testicular tissue from stallions of different ages (prepubertal, young, mature and old). Gene and protein expression were assessed by real-time quantitative polymerase chain reaction (real-time qPCR), immunohistochemistry and multiple immunofluorescence labelling. There were no differences in LHR mRNA expression in epididymal and testicular parenchyma in stallions of different age. In contrast, expression of FSHR and CYP19A1 in caput, corpus and cauda epididymis and in testicular parenchyma increased with age (P < 0.001). Immunolabelling for LHR, FSHR and aromatase was influenced by puberty. In postpubertal stallions, positive staining for LHR and aromatase was detected in Leydig cells, whereas protein expression of FSHR was present in Sertoli cells and primary spermatocytes. In prepubertal colts, staining for LHR, FSHR and aromatase was detected in seminiferous tubules. In epididymal tissue, aromatase was present in the cauda epididymis only, regardless of age. In conclusion, the results highlight the significance of gonadotropin action and oestrogen production for the maturation of male reproductive tissue in the horse. The presence of FSHR in the seminiferous tubules suggests effects of FSH on spermatogenesis in this species. The importance of oestrogen production for maintenance of testicular function in stallions was confirmed.
Collapse
|
8
|
Kaneko Y, Dailey T, Weinbren NL, Rizzi J, Tamboli C, Allickson JG, Kuzmin-Nichols N, Sanberg PR, Eve DJ, Tajiri N, Borlongan CV. The battle of the sexes for stroke therapy: female- versus male-derived stem cells. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2014; 12:405-412. [PMID: 23469849 DOI: 10.2174/1871527311312030013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Revised: 09/10/2012] [Accepted: 09/14/2012] [Indexed: 12/14/2022]
Abstract
Cell therapy is a major discipline of regenerative medicine that has been continually growing over the last two decades. The aging of the population necessitates discovery of therapeutic innovations to combat debilitating disorders, such as stroke. Menstrual blood and Sertoli cells are two gender-specific sources of viable transplantable cells for stroke therapy. The use of autologous cells for the subacute phase of stroke offers practical clinical application. Menstrual blood cells are readily available, display proliferative capacity, pluripotency and angiogenic features, and, following transplantation in stroke models, have the ability to migrate to the infarct site, regulate the inflammatory response, secrete neurotrophic factors, and have the possibility to differentiate into neural lineage. Similarly, the testis-derived Sertoli cells secrete many growth and trophic factors, are highly immunosuppressive, and exert neuroprotective effects in animal models of neurological disorders. We highlight the practicality of experimental and clinical application of menstrual blood cells and Sertoli cells to treat stroke, from cell isolation and cryopreservation to administration.
Collapse
Affiliation(s)
- Yuji Kaneko
- Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| | - Travis Dailey
- Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| | - Nathan L Weinbren
- Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| | - Jessica Rizzi
- Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| | - Cyrus Tamboli
- Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| | | | | | - Paul R Sanberg
- Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| | - David J Eve
- Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| | - Naoki Tajiri
- Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| | - Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| |
Collapse
|
9
|
Albert S, Wistuba J, Eildermann K, Ehmcke J, Schlatt S, Gromoll J, Kossack N. Comparative Marker Analysis after Isolation and Culture of Testicular Cells from the Immature Marmoset. Cells Tissues Organs 2012; 196:543-54. [DOI: 10.1159/000339010] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/24/2012] [Indexed: 11/19/2022] Open
|
10
|
Chui K, Trivedi A, Cheng CY, Cherbavaz DB, Dazin PF, Huynh ALT, Mitchell JB, Rabinovich GA, Noble-Haeusslein LJ, John CM. Characterization and functionality of proliferative human Sertoli cells. Cell Transplant 2010; 20:619-35. [PMID: 21054948 DOI: 10.3727/096368910x536563] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
It has long been thought that mammalian Sertoli cells are terminally differentiated and nondividing postpuberty. For most previous in vitro studies immature rodent testes have been the source of Sertoli cells and these have shown little proliferative ability when cultured. We have isolated and characterized Sertoli cells from human cadaveric testes from seven donors ranging from 12 to 36 years of age. The cells proliferated readily in vitro under the optimized conditions used with a doubling time of approximately 4 days. Nuclear 5-ethynyl-2'-deoxyuridine (EdU) incorporation confirmed that dividing cells represented the majority of the population. Classical Sertoli cell ultrastructural features, lipid droplet accumulation, and immunoexpression of GATA-4, Sox9, and the FSH receptor (FSHr) were observed by electron and fluorescence microscopy, respectively. Flow cytometry revealed the expression of GATA-4 and Sox9 by more than 99% of the cells, and abundant expression of a number of markers indicative of multipotent mesenchymal cells. Low detection of endogenous alkaline phosphatase activity after passaging showed that few peritubular myoid cells were present. GATA-4 and SOX9 expression were confirmed by reverse transcription polymerase chain reaction (RT-PCR), along with expression of stem cell factor (SCF), glial cell line-derived neurotrophic factor (GDNF), and bone morphogenic protein 4 (BMP4). Tight junctions were formed by Sertoli cells plated on transwell inserts coated with fibronectin as revealed by increased transepithelial electrical resistance (TER) and polarized secretion of the immunoregulatory protein, galectin-1. These primary Sertoli cell populations could be expanded dramatically in vitro and could be cryopreserved. The results show that functional human Sertoli cells can be propagated in vitro from testicular cells isolated from adult testis. The proliferative human Sertoli cells should have important applications in studying infertility, reproductive toxicology, testicular cancer, and spermatogenesis, and due to their unique biological properties potentially could be useful in cell therapy.
Collapse
Affiliation(s)
- Kitty Chui
- MandalMed, Inc., San Francisco, CA 94107, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Radu A, Pichon C, Camparo P, Antoine M, Allory Y, Couvelard A, Fromont G, Hai MTV, Ghinea N. Expression of follicle-stimulating hormone receptor in tumor blood vessels. N Engl J Med 2010; 363:1621-30. [PMID: 20961245 DOI: 10.1056/nejmoa1001283] [Citation(s) in RCA: 222] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND In adult humans, the follicle-stimulating hormone (FSH) receptor is expressed only in the granulosa cells of the ovary and the Sertoli cells of the testis. It is minimally expressed by the endothelial cells of gonadal blood vessels. METHODS We used immunohistochemical and immunoblotting techniques involving four separate FSH-receptor-specific monoclonal antibodies that recognize different FSH receptor epitopes and in situ hybridization to detect FSH receptor in tissue samples from patients with a wide range of tumors. Immunoelectron microscopy was used to detect FSH receptor in mouse tumors. RESULTS In all 1336 patients examined, FSH receptor was expressed by endothelial cells in tumors of all grades, including early T1 tumors. The tumors were located in the prostate, breast, colon, pancreas, urinary bladder, kidney, lung, liver, stomach, testis, and ovary. In specimens obtained during surgery performed to remove tumors, the FSH receptor was not expressed in the normal tissues located more than 10 mm from the tumors. The tumor lymphatic vessels did not express FSH receptor. The endothelial cells that expressed FSH receptor were located at the periphery of the tumors in a layer that was approximately 10 mm thick; this layer extended both into and outside of the tumor. Immunoelectron microscopy in mice with xenograft tumors, after perfusion with anti–FSH-receptor antibodies coupled to colloidal gold, showed that the FSH receptor is exposed on the luminal endothelial surface and can bind and internalize circulating ligands. CONCLUSIONS FSH receptor is selectively expressed on the surface of the blood vessels of a wide range of tumors. (Funded by INSERM.).
Collapse
|
12
|
Segretain D, Gilleron J, Carette D, Denizot JP, Pointis G. Differential time course of FSH/FSH receptor complex endocytosis within sertoli and germ cells during rat testis development. Dev Dyn 2010; 239:1113-23. [PMID: 20201104 DOI: 10.1002/dvdy.22261] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Follicle-stimulating hormone (FSH) is required for initiation and maintenance of spermatogenesis, a dynamic process of cell proliferation and maturation. By using FSH-gold particles and pulse-chase experiments, we analyzed the kinetics of FSH endocytosis in Sertoli and germ cells during development. Ultrastructural time-dependent analysis demonstrates that FSH was first located on plasma membrane, before being accumulated within the endosomal compartment, in the early endosomes, identified by morphological criteria and Rab-5 colocalization. Thereafter, FSH-gold was routed to the degradation pathway. The FSH endocytosis kinetic was similar in Sertoli cells, spermatogonia and spermatocytes. However, quantitative analysis of gold particles revealed differences in the dynamic of FSH accumulation in the endosomes between immature and mature rats. This age-dependent kinetic of FSH endocytosis, mostly detectable by ultrastructural analysis associated with quantitative data, argues for a potential new regulatory mechanism of the FSH signalling pathway that could occur during maturation of testicular cells.
Collapse
|
13
|
Abstract
Follicle stimulating hormone (FSH) is a glycoprotein hormone secreted by the pituitary gland that, together with luteinizing hormone (LH), controls development, maturation and function of the gonad. Like the related hormones, LH, thyroid stimulating hormone (TSH) and human chorionic gonadotropin (hCG), FSH consists of two polypeptide chains, α and β, bearing carbohydrate moietiesN-linked to asparagine (Asn) residues. The α subunit is common to all members of the glycoprotein hormone family, whereas the β subunit, although structurally very similar, differs in each hormone and confers specificity of action.
Collapse
|
14
|
Foresta C, Selice R, Moretti A, Pati MA, Carraro M, Engl B, Garolla A. Gonadotropin administration after gonadotropin-releasing-hormone agonist: a therapeutic option in severe testiculopathies. Fertil Steril 2008; 92:1326-1332. [PMID: 18829015 DOI: 10.1016/j.fertnstert.2008.07.1766] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2008] [Revised: 07/24/2008] [Accepted: 07/24/2008] [Indexed: 10/21/2022]
Abstract
OBJECTIVE To evaluate the effect of recombinant human follicle-stimulating hormone (FSH) plus human chorionic gonadotropin (hCG) on seminal parameters and pregnancy rate in severe testiculopathies after high FSH plasma concentrations have been suppressed. DESIGN Prospective, controlled, randomized clinical study. SETTING Infertility center at a university hospital. PATIENT(S) Eighty-seven men affected by severe testiculopathy. INTERVENTION(S) We treated 57 men with a gonadotropin-releasing hormone agonist (GnRH-a) and then with recombinant human FSH and hCG, and 30 patients did not receive any treatment. Seminal parameters and sperm aneuploidies were evaluated during and after the treatment period. Couples did not achieve a spontaneous pregnancy received assisted reproduction treatment. MAIN OUTCOME MEASURE(S) Seminal parameters, sperm aneuploidies, testicular cytologic analysis, FSH, luteinizing hormone, testosterone, inhibin B concentrations, and pregnancy rate. RESULT(S) After the therapy period, the treated group showed statistically significant improvement in sperm parameters and sperm aneuploidies. No changes were observed in the untreated group. A trend toward an increase in pregnancy rate also was observed among treated couples (cumulative pregnancy rates 31.6% treated vs. 20.0% untreated), although the increase was not statistically significant. The improvement of seminal parameters in the treated group allowed some patients to undergo in vitro fertilization-embryo transfer instead of intracytoplasmic sperm injection. CONCLUSION(S) Results from this controlled, randomized clinical trial show that FSH therapy improves sperm parameters in severe male factor infertility when endogenous high FSH plasma levels are suppressed. In cases of severely impaired testicles, a rational treatment of male infertility is mandatory to improve the outcome of assisted reproduction techniques.
Collapse
Affiliation(s)
- Carlo Foresta
- Section of Clinical Pathology and Centre for Male Gamete Cryopreservation, Department of Histology, Microbiology, and Medical Biotechnologies, University of Padova, Padova, Italy.
| | - Riccardo Selice
- Section of Clinical Pathology and Centre for Male Gamete Cryopreservation, Department of Histology, Microbiology, and Medical Biotechnologies, University of Padova, Padova, Italy
| | - Afra Moretti
- Section of Clinical Pathology and Centre for Male Gamete Cryopreservation, Department of Histology, Microbiology, and Medical Biotechnologies, University of Padova, Padova, Italy
| | - Mauro Antonio Pati
- Section of Clinical Pathology and Centre for Male Gamete Cryopreservation, Department of Histology, Microbiology, and Medical Biotechnologies, University of Padova, Padova, Italy
| | - Marina Carraro
- Section of Clinical Pathology and Centre for Male Gamete Cryopreservation, Department of Histology, Microbiology, and Medical Biotechnologies, University of Padova, Padova, Italy
| | - Bruno Engl
- Obstetrics and Gynaecology Unit, Brunico, Italy
| | - Andrea Garolla
- Section of Clinical Pathology and Centre for Male Gamete Cryopreservation, Department of Histology, Microbiology, and Medical Biotechnologies, University of Padova, Padova, Italy
| |
Collapse
|
15
|
Sharma CS, Sarkar S, Periyakaruppan A, Ravichandran P, Sadanandan B, Ramesh V, Thomas R, Hall JC, Wilson BL, Ramesh GT. Simulated microgravity activates apoptosis and NF-kappaB in mice testis. Mol Cell Biochem 2008; 313:71-8. [PMID: 18385949 DOI: 10.1007/s11010-008-9743-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2008] [Accepted: 03/13/2008] [Indexed: 10/22/2022]
Abstract
Microgravity is known to have significant effect on all aspects of reproductive function in animal models. Recent studies have also shown that microgravity induces changes at the cellular level, including apoptosis. Our effort here was to study the effect of simulated microgravity on caspase-8 and the caspase-3 activities, the effectors of the apoptotic pathway and on the transcription factor NF-kappaB a signaling molecule in mouse testis. Morey-Holton hind limb suspension model was used to simulate microgravity. Caspase-8 and 3 fluorometric assays were carried out and HLS mice testis exhibited a 51% increase in caspase-8 and caspase-3 compared to the controls. A sandwich ELISA-based immunoassay was carried out for detection of NF-kappaB which again significantly increased in the test mice. Testosterone levels were measured using an ELISA kit and in HLS mice the decrease was significant. There was also a significant decrease in testis weight in the test mice. Simulated microgravity activates caspase 8, 3 and NF-kappaB necessary to stimulate the apoptotic pathway in mice testis. This may account for the drop in testis weight and testosterone level further affecting testicular physiology and function.
Collapse
Affiliation(s)
- Chidananda S Sharma
- Department of Biology, Molecular Toxicology Laboratory, Center for Biotechnology & Biomedical Sciences, Norfolk State University, Norfolk, VA 23504, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
|
17
|
Hermann BP, Heckert LL. Transcriptional regulation of the FSH receptor: new perspectives. Mol Cell Endocrinol 2007; 260-262:100-8. [PMID: 17084019 PMCID: PMC3682414 DOI: 10.1016/j.mce.2006.09.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2005] [Accepted: 11/30/2005] [Indexed: 02/08/2023]
Abstract
The cell-surface receptor for the gonadotropin follicle-stimulating hormone (FSH) is expressed exclusively on Sertoli cells of the testis and granulosa cells of the ovary. FSH signal transduction through its receptor (Fshr) is critical for the timing and maintenance of normal gametogenesis in the mammalian gonad. In the 13 years since the gene encoding Fshr was first cloned, the mechanisms controlling its transcription have been extensively examined, but a clear understanding of what drives its unique cell-specificity remains elusive. Current knowledge of basal Fshr transcription highlights the role of an E-box in the proximal promoter which is bound by the basic helix-loop-helix transcription factors upstream stimulatory factor 1 (Usf1) and Usf2. Recent studies utilizing knockout mice and chromatin immunoprecipitation validated the importance of Usf to Fshr transcription and demonstrated a sexually dimorphic requirement for the Usf proteins to maintain normal Fshr expression. Studies have also shown that the promoter region itself is insufficient for appropriate Fshr expression in transgenic mice, indicating Fshr transcription depends on regulatory elements that lie outside of the promoter. Identification of such elements has been propelled by recent availability of genome sequence data, which facilitated studies using comparative genomics, DNase I hypersensitivity mapping, and transgenic analysis with large fragments of DNA. This review will focus on the current understanding of transcriptional regulatory processes that control expression of rat Fshr, including recent advances from our laboratory.
Collapse
Affiliation(s)
| | - Leslie L. Heckert
- Corresponding Author: Address: Department of Molecular and Integrative Physiology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160 USA Telephone: 913-588-7488 Fax: 913-588-7430
| |
Collapse
|
18
|
Grover A, Smith CE, Gregory M, Cyr DG, Sairam MR, Hermo L. Effects of FSH receptor deletion on epididymal tubules and sperm morphology, numbers, and motility. Mol Reprod Dev 2005; 72:135-44. [PMID: 15973687 DOI: 10.1002/mrd.20303] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Follicle stimulating hormone (FSH) interacts with its cognate receptor (R) on Sertoli cells within the testis and plays an important role in the maintenance of spermatogenesis. Male FSH-R knockout (FORKO) mice show fewer Sertoli cells and many that are structurally abnormal and as a consequence fewer germ cells. Lower levels of serum testosterone (T) and androgen binding protein (ABP) also occur, along with reduced fertility. To assess the effects of FSH-R depletion as an outcome of testicular abnormalities, sperm from the cauda epididymidis were counted and examined ultrastructurally. As reduced fertility may also reflect changes to the epididymis, the secondary responses of the epididymis to lower T and ABP levels were also examined by comparing differences in sizes of epididymal tubules in various regions of FORKO and wild type (WT) mice. Sperm motility was evaluated in FORKO mice and compared to that of WT mice by computer assisted sperm analysis (CASA). Quantitatively, the data revealed that epithelial areas of the caput and corpus epididymidis were significantly smaller in FORKO mice compared to WT mice. Cauda epididymal sperm counts in FORKO mice were also much lower than in WT mice. This resulted in changes to 9 out of 14 sperm motility parameters, related mostly to velocity measures, which were significantly lower in the FORKO mice. The greatest change was observed relative to the percent static sperm, which was elevated by 20% in FORKO mice compared to controls. EM analyses revealed major changes to the structure of the heads and tails of cauda luminal sperm in FORKO mice. Taken together these data suggest a key role for the FSH receptor in maintaining Sertoli cells to sustain normal sperm numbers and proper shapes of their heads and tails. In addition, the shrinkage in epididymal epithelial areas observed in FORKO mice likely reflect direct and/or indirect changes in the functions of these cells and their role in promoting sperm motility, which is noticeably altered in FORKO mice.
Collapse
Affiliation(s)
- Amit Grover
- Molecular Reproduction Research Laboratory, Clinical Research Institute of Montreal, 110 Pine Avenue West, Montreal, Quebec, Canada H2W 1R7
| | | | | | | | | | | |
Collapse
|
19
|
Steger K. Possible predictive factors for ICSI? Molecular biology techniques in combination with therapeutic testicular biopsies. Andrologia 2003; 35:200-8. [PMID: 12950403 DOI: 10.1046/j.1439-0272.2003.00581.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Applying intracytoplasmic sperm injection (ICSI), the selection of an unsuccessful spermatozoon results in great emotional consequences for the couple. Therefore, there is a need for a prognostic parameter to estimate their chances for successful fertility treatment. This review summarizes both the main reasons for spermatogenic impairment, and possible predictive factors for successful sperm retrieval applying testicular sperm extraction and outcome of ICSI. While basic sperm parameters, aetiology and type of spermatozoa, and serum follicle-stimulating hormone and inhibin levels have been shown to be unrelated to the outcome of ICSI, Y-chromosome microdeletions are known to have a negative influence on the fertilizing capacity of spermatozoa. Recently, a significant correlation has been reported between the protamine-1 to protamine-2 mRNA ratio in haploid spermatids of testicular biopsies and the ability of spermatozoa for successful fertilization of an oocyte. In future, both the outstanding role of the haploid spermatids and the involvement of molecular biological techniques will improve the role of therapeutic testicular biopsies.
Collapse
Affiliation(s)
- K Steger
- Institute of Veterinary Anatomy, Histology and Embryology, Giessen, Germany.
| |
Collapse
|
20
|
Tesarik J, Martinez F, Rienzi L, Iacobelli M, Ubaldi F, Mendoza C, Greco E. In-vitro effects of FSH and testosterone withdrawal on caspase activation and DNA fragmentation in different cell types of human seminiferous epithelium. Hum Reprod 2002; 17:1811-9. [PMID: 12093844 DOI: 10.1093/humrep/17.7.1811] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Caspases are downstream elements of apoptosis-mediating pathways initiated by the Fas ligand/Fas receptor system which is supposed to play a central role in the regulation of apoptosis in the human seminiferous epithelium. However, caspase activity in different cell types of this epithelium has never been addressed. METHODS AND RESULTS We evaluated caspase activity and DNA integrity in Sertoli and germ cells within in-vitro cultured segments of human seminiferous tubules after induction of apoptosis by FSH or testosterone withdrawal. FSH withdrawal increased the incidence of DNA fragmentation in meiotic (primary spermatocytes) and post-meiotic (spermatids) germ cells without producing any detectable effect on caspase activity in these cells and without affecting DNA integrity or caspase activity in Sertoli cells. Testosterone withdrawal stimulated caspase activity and produced DNA fragmentation in Sertoli cells, but showed only a weak effect on DNA fragmentation in germ cells and did not alter germ cell caspase activity. CONCLUSIONS These findings confirm the central role of caspases in apoptosis of Sertoli cells. However, they also suggest that acute apoptosis of germ cells in the adult human testis occurs in a caspase-independent way and is controlled by Sertoli cells via an as yet undetermined mechanism.
Collapse
Affiliation(s)
- Jan Tesarik
- Molecular Assisted Reproduction & Genetics, Gracia 36, 18002 Granada, Spain
| | | | | | | | | | | | | |
Collapse
|
21
|
Nakayama Y, Yamamoto T, Oba Y, Nagahama Y, Abé S. Molecular cloning, functional characterization, and gene expression of a follicle-stimulating hormone receptor in the testis of newt Cynops pyrrhogaster. Biochem Biophys Res Commun 2000; 275:121-8. [PMID: 10944452 DOI: 10.1006/bbrc.2000.3253] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We previously demonstrated in vitro that mammalian follicle-stimulating hormone (FSH) stimulates the proliferation of newt secondary spermatogonia and their differentiation into primary spermatocytes. In the current study, we isolated a cDNA from newt testis that encodes a FSH receptor (FSH-R). The total sequence homology in the deduced protein of the newt was approximately 70% with mammalian FSH-Rs. Mammalian cells, transiently transfected with the cloned newt FSH-R cDNA, displayed specific binding to [(125)I] human FSH and cAMP accumulation, indicating that the cloned cDNA encodes a functional newt FSH-R protein. Northern blot analysis revealed a single transcript of approximately 3.0 kb length that was synthesized in testicular somatic cells (mainly Sertoli cells) from spermatogonial to spermatid stages with the highest level expressed during the primary spermatocytes stage. These results demonstrate that FSH stimulates newt spermatogenesis through the FSH-R. This study, as far as we know, reports for the first time the cloning of an amphibian FSH-R cDNA.
Collapse
Affiliation(s)
- Y Nakayama
- Kumamoto University, Kumamoto, 860-8555, Japan
| | | | | | | | | |
Collapse
|
22
|
Bar-Shira Maymon B, Paz G, Elliott DJ, Hammel I, Kleiman SE, Yogev L, Hauser R, Botchan A, Yavetz H. Maturation phenotype of Sertoli cells in testicular biopsies of azoospermic men. Hum Reprod 2000; 15:1537-42. [PMID: 10875862 DOI: 10.1093/humrep/15.7.1537] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The involvement of Sertoli cells in different spermatogenic impairments has been studied by an immunohistomorphometric technique using cytokeratin-18 (CK-18) as a marker for immature Sertoli cells. CK-18 is known to be expressed in Sertoli cells during prenatal and prepubertal differentiation and is normally lost at puberty. Forty-nine azoospermic men were included in the current study. Quantitative measurements on testicular biopsies revealed the highest CK-18 expression in the mixed atrophy biopsies (22 men), a lower expression in the Sertoli cell-only (SCO) biopsies (12 men), and minimal residual staining in the group considered as representing normal spermatogenesis (six obstructive azoospermia patients). The cytokeratin immunopositive-stained tubules were associated either with arrest in spermatogenesis or with SCO. Examination of sections from nine men with microdeletions in the AZF region of the Y chromosome revealed that these men were either negative for CK-18 expression or showed only weak residual staining. This may suggest that the spermatogenic defect in the AZF-deleted men originates in the germ cell and has no impact on Sertoli cell maturation. The cause that determined the spermatogenic defect in the other cases of male infertility with high CK-18 expression may have damaged both the Sertoli and the germ cells.
Collapse
Affiliation(s)
- B Bar-Shira Maymon
- Institute for the Study of Fertility, Lis Maternity Hospital, Tel Aviv Sourasky Medical Center, Israel.
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Meehan T, Schlatt S, O'Bryan MK, de Kretser DM, Loveland KL. Regulation of germ cell and Sertoli cell development by activin, follistatin, and FSH. Dev Biol 2000; 220:225-37. [PMID: 10753512 DOI: 10.1006/dbio.2000.9625] [Citation(s) in RCA: 133] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have demonstrated a role for activin A, follistatin, and FSH in male germ cell differentiation at the time when spermatogonial stem cells and committed spermatogonia first appear in the developing testis. Testis fragments from 3-day-old rats were cultured for 1 or 3 days with various combinations of these factors, incubated with bromodeoxyuridine (BrdU) to label proliferating cells, and then processed for stereological analysis and detection of BrdU incorporation. Gonocyte numbers were significantly elevated in cultures treated with activin, while the combination of FSH and the activin antagonist, follistatin, increased the proportion of spermatogonia in the germ cell population after 3 days. All fragment groups treated with FSH contained a significantly higher proportion of proliferating Sertoli cells, while activin and follistatin each reduced Sertoli cell division. In situ hybridization and immunohistochemistry on normal rat testes demonstrated that gonocytes, but not spermatogonia, contain the activin beta(A) subunit mRNA and protein. In contrast, gonocytes first expressed follistatin mRNA and protein at 3 days after birth, concordant with the transition of gonocytes to spermatogonia. Collectively, these data demonstrate that germ cells have the potential to regulate their own maturation through production of endogenous activin A and follistatin. Sertoli cells were observed to produce the activin/inhibin beta(A) subunit, the inhibin alpha subunit, and follistatin, demonstrating that these cells have the potential to regulate germ cell maturation as well as their own development. These findings indicate that local regulation of activin bioactivity may underpin the coordinated development of germ cells and somatic cells at the onset of spermatogenesis.
Collapse
Affiliation(s)
- T Meehan
- Monash Institute of Reproduction & Development, Monash Medical Centre, Clayton, Victoria, 3168, Australia
| | | | | | | | | |
Collapse
|
24
|
Foresta C, Bettella A, Merico M, Garolla A, Plebani M, Ferlin A, Rossato M. FSH in the treatment of oligozoospermia. Mol Cell Endocrinol 2000; 161:89-97. [PMID: 10773396 DOI: 10.1016/s0303-7207(99)00228-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The aim of this study was to individuate parameters able to distinguish oligozoospermic subjects who will respond to follicle-stimulating hormone (FSH) therapy. A group of 135 oligozoospermic subjects was divided in three groups considering basal FSH and inhibin B concentrations: group A (normal FSH and inhibin B) characterized by moderate hypospermatogenesis sometimes associated to partial spermatidic arrest; group B (high FSH and normal inhibin B) characterized by hypospermatogenesis associated or not to spermatogonial/spermatocytic arrest; group C (high FSH and low inhibin B) characterized by severe hypospermatogenesis. Seventy-eight patients were treated with FSH at the dose of 75 IU on alternate days while 57 were treated with the same dose every day for 3 months. After FSH treatment a significant increase in ejaculated sperm concentration was observed only in oligozoospermic subjects with normal basal FSH and inhibin B plasma levels (group A) showing a testicular cytological picture of moderate hypospermatogenesis. In these subjects no differences in sperm production were observed between the two protocols of therapy. In the remaining patients of group A, characterized by hypospermatogenesis associated with maturation arrest at spermatidic level and in group B and C, no increase in sperm concentration was observed after therapy. These data suggest that FSH treatment may have a role in oligozoospermic subjects only when the spermatogenetic alterations consist in germ cell depopulation without maturative disturbances and with normal FSH concentrations.
Collapse
Affiliation(s)
- C Foresta
- Department of Medical and Surgical Sciences, University of Padova, Clinica Medica 3, Via Ospedale 105, 35128, Padova, Italy.
| | | | | | | | | | | | | |
Collapse
|
25
|
Baccetti B, Collodel G, Costantino-Ceccarini E, Eshkol A, Gambera L, Moretti E, Strazza M, Piomboni P. Localization of human follicle-stimulating hormone in the testis. FASEB J 1998; 12:1045-54. [PMID: 9707177 DOI: 10.1096/fasebj.12.11.1045] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Localization of the follicle-stimulating hormone (FSH) molecule and its receptor (FSHR), as well as the role of FSH in Sertoli cell mitosis and maturation, has been demonstrated by several investigators in human and murine testis by detecting the localization of anti-FSH antibodies or [(131)I]-labeled FSH and by detecting FSH receptor (FSHR) mRNA by in situ hybridization, or FSHR by anti-FSHR antibodies. The presence of FSH in germinal cells is controversial or, in humans, excluded. We have investigated the distribution of the human FSH molecule and its receptor in human and mouse testicular cells under different experimental conditions, at the submicroscopical level, by using a better antigenicity conservative procedure. Thus, the distribution of FSH and of the messenger RNA for its receptor in Sertoli cells has now been clarified. In germinal cells, our observations demonstrate the presence of FSH and the FSHR mRNA: the first on the plasma membrane and in endocytotic vesicles, and the second scattered in the cytoplasm. The cells presenting the higher amount of positivity ranged from spermatogonia to spermatocytes, including round spermatids. Penetration was by the endocytosis via membrane vesicles in which the FSHR is present, whereas its messenger is largely present in the cytoplasm and is responsible for the binding and subsequent internalization of the FSH molecule. As a control, human FSH was administered in vitro to the Y1 mouse cell line, which was stably transfected with cDNA for FSHR and devoid of endogenous FSH. The FSH molecule has been localized by monoclonal antibodies on plasma membranes and vesicles, and the FSHR mRNA was found scattered in the cytoplasm after in situ hybridization. We can now conclude that FSH is present in Sertoli cells and in round germinal cells, both expressing the FSHR. FSH penetrates in a similar way in both kinds of cells via endocytosis, and is therefore subsequently localized in the same membranous organelles.
Collapse
Affiliation(s)
- B Baccetti
- Institute of General Biology of the University of Siena and Center for the Study of Germinal Cells, CNR, Italy
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Foresta C, Bettella A, Ferlin A, Garolla A, Rossato M. Evidence for a stimulatory role of follicle-stimulating hormone on the spermatogonial population in adult males. Fertil Steril 1998; 69:636-42. [PMID: 9548151 DOI: 10.1016/s0015-0282(98)00008-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To evaluate the effects of treatment with FSH on seminal indices and on the seminiferous epithelium of oligozoospermic subjects with normal FSH plasma levels. DESIGN Placebo-controlled, double-blind randomized study. SETTING Academic setting. PATIENT(S) Ninety subjects with idiopathic oligozoospermia (sperm count of < 10 x 10(6)/mL) and normal plasma levels of FSH. INTERVENTION(S) Three months of treatment with FSH (60 patients) or placebo (30 patients); bilateral testicular fine-needle aspiration. MAIN OUTCOME MEASURE(S) Seminal indices; testicular cytologic features; plasma levels of FSH, LH, and testosterone; and ultrasonographic testicular examination. RESULT(S) According to seminal indices, patients treated with FSH and placebo were classified as nonresponders or as responders (as determined by at least a doubling of sperm count). No placebo-treated patients responded to treatment. Among FSH-treated patients, 20 responded to hormonal treatment and 40 did not. The results of pretreatment cytologic examination of testicular specimens from patients who did not respond to FSH treatment were consistent with hypospermatogenesis associated with maturational disturbances at the spermatid level. In contrast, patients who responded to treatment with FSH had isolated hypospermatogenesis without maturational disturbances. After FSH therapy, we detected an increase of spermatogonia and spermatocyte population in both the responder and nonresponder subjects. This increase was associated with an activation of spermatogenic and spermiogenic processes and with a rise in ejaculated sperm concentration only when isolated hypospermatogenesis was present (responder patients). CONCLUSION(S) The findings of this study demonstrate that FSH treatment increases the spermatogonial population in men. This treatment may be appropriate for oligozoospermic subjects who have normal FSH plasma levels and a testicular evaluation characterized by hypospermatogenesis without maturational disturbances.
Collapse
Affiliation(s)
- C Foresta
- Clinica Medica 3, University of Padova, Italy.
| | | | | | | | | |
Collapse
|
27
|
Simoni M, Gromoll J, Nieschlag E. The follicle-stimulating hormone receptor: biochemistry, molecular biology, physiology, and pathophysiology. Endocr Rev 1997; 18:739-73. [PMID: 9408742 DOI: 10.1210/edrv.18.6.0320] [Citation(s) in RCA: 182] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- M Simoni
- Institute of Reproductive Medicine of the University, Münster, Germany
| | | | | |
Collapse
|
28
|
Weinbauer GF, Nieschlag E. Endocrine control of germ cell proliferation in the primate testis. What do we really know? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1997; 424:51-8. [PMID: 9361762 DOI: 10.1007/978-1-4615-5913-9_6] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The present chapter reviews current knowledge concerning hormonal regulation of gametogenesis in the primate testis. LH/testosterone and FSH are the prime regulators of primate spermatogenesis. Although either hormone is capable of stimulating all phases of the spermatogenic process including the formation of sperm, the combination of both hormones is necessary in most instances to achieve quantitatively normal germ cell numbers. Sertoli cell proliferation can also be induced by either hormone in juvenile monkeys. Evidence for differential effects of testosterone and FSH on gametogenesis, however, is lacking and a synergistic effect is observed when they are combined. Receptors for androgens and FSH occur exclusively on testicular somatic cells and, hence, the trophic effects of these hormones on germ cell numbers are indirect ones. Interestingly, both hormones seem to have a common target, the spermatogonial population but it is unknown how such an indirect albeit highly specific effect is mediated. Whether the trophic hormone action influences germ cell numbers via increased proliferation or decreased cell death or both remains to be seen. There is evidence to suggest that the local androgen requirements for primate spermatogenesis might be comparatively high.
Collapse
Affiliation(s)
- G F Weinbauer
- Institute of Reproductive Medicine of the University, Münster, Germany
| | | |
Collapse
|