1
|
Bodu M, Hitit M, Memili E. Harnessing the value of fertility biomarkers in bull sperm for buck sperm. Anim Reprod Sci 2025; 272:107643. [PMID: 39577268 DOI: 10.1016/j.anireprosci.2024.107643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 10/14/2024] [Accepted: 11/12/2024] [Indexed: 11/24/2024]
Abstract
Efficient and sustainable reproduction and production of cattle and goats are vitally important for ensuring global food security. There is a need for potent biomarkers to accurately evaluate semen quality and predict male fertility. Although there is a reasonable set of biomarkers identified in bull sperm, there is a significant lack of such information in buck sperm along with a lack of transfer of proven technologies in goat reproductive biotechnology. These gaps are important problems because they are preventing advances in fundamental andrology and applied science of goat production. Both cattle and goats are ruminants, and they share significant similarities in their genetics and physiology although subtle differences do exist. This review harnesses the power of utilizing the knowledge developed in bull sperm to generate information on buck sperm fertility markers. These include genomic, functional genomic, epigenomic fertility markers. Revealing molecular underpinnings of such similarity and diversity using systems biology is expected to advance both fundamental and applied andrology of livestock and endangered species.
Collapse
Affiliation(s)
- Mustafa Bodu
- College of Agriculture, Food and Natural Resources, Cooperative Agricultural Research Center, Prairie View A&M University, Prairie View, TX, United States; Department of Reproduction and Artificial Insemination, Faculty of Veterinary Medicine, Selcuk University, Konya, Türkiye.
| | - Mustafa Hitit
- College of Agriculture, Food and Natural Resources, Cooperative Agricultural Research Center, Prairie View A&M University, Prairie View, TX, United States.
| | - Erdogan Memili
- College of Agriculture, Food and Natural Resources, Cooperative Agricultural Research Center, Prairie View A&M University, Prairie View, TX, United States.
| |
Collapse
|
2
|
Tabęcka-Łonczyńska A, Koszła O, Sołek P. Unraveling the anti-androgenic mechanism of tris(2,3-dibromopropyl) isocyanurate (TBC) via the non-classical testosterone pathway and steroidogenesis: Potential human reproductive health implications. CHEMOSPHERE 2024; 363:142802. [PMID: 38996977 DOI: 10.1016/j.chemosphere.2024.142802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 07/04/2024] [Accepted: 07/06/2024] [Indexed: 07/14/2024]
Abstract
The decline in male reproductive health, characterized by diminishing sperm count and testosterone levels, has raised concerns about environmental influences, particularly endocrine-disrupting chemicals (EDCs). Tris(2,3-dibromopropyl)isocyanurate (TBC), a novel brominated flame retardant widely used in electronics, textiles, and furniture, has emerged as a significant environmental contaminant with potential reproductive health implications. In this study, we investigated the molecular mechanisms underlying TBC-induced reproductive toxicity, particularly focusing on its impact on steroidogenesis and androgen signaling pathways using the GC-1 spg cell line as an in vitro model. Exposure of GC-1 spg cells to TBC, alone or in combination with testosterone or the anti-androgen flutamide resulted in decreased metabolic activity and increased lactate dehydrogenase release, indicating cytotoxic effects. Furthermore, TBC exposure led to a reduction in progesterone synthesis, while testosterone production remained unaffected. Interestingly, estradiol synthesis was diminished after TBC exposure, suggesting a disruption in steroid hormone balance critical for spermatogenesis. Mechanistic investigations revealed alterations in key proteins involved in the non-classical testosterone pathway and steroidogenesis. TBC exposure downregulated epidermal growth factor receptor (EGFR), protein kinase B (AKT), and phosphorylated cyclic AMP response element-binding protein (p-CREB), indicating suppression of non-classical androgen signaling. Additionally, decreased levels of steroidogenic acute regulatory protein (StAR) and 3-beta-hydroxysteroid dehydrogenase (HSD3β1) suggest impaired steroidogenesis. Here we uncover the intricate molecular mechanisms underlying TBC-induced reproductive toxicity, highlighting its potential to disrupt steroid hormone synthesis and androgen signaling pathways. Understanding the adverse effects of TBC on male reproductive health is crucial for developing strategies to mitigate its environmental impact and safeguard human fertility.
Collapse
Affiliation(s)
- Anna Tabęcka-Łonczyńska
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225, Rzeszów, Poland.
| | - Oliwia Koszła
- Department of Biopharmacy, Medical University of Lublin, Chodźki 4a, 20-093, Lublin, Poland.
| | - Przemysław Sołek
- Department of Biopharmacy, Medical University of Lublin, Chodźki 4a, 20-093, Lublin, Poland; Department of Biochemistry and Toxicology, University of Life Sciences, Akademicka 13, 20-950, Lublin, Poland.
| |
Collapse
|
3
|
Li L, Lin W, Wang Z, Huang R, Xia H, Li Z, Deng J, Ye T, Huang Y, Yang Y. Hormone Regulation in Testicular Development and Function. Int J Mol Sci 2024; 25:5805. [PMID: 38891991 PMCID: PMC11172568 DOI: 10.3390/ijms25115805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 05/01/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
The testes serve as the primary source of androgens and the site of spermatogenesis, with their development and function governed by hormonal actions via endocrine and paracrine pathways. Male fertility hinges on the availability of testosterone, a cornerstone of spermatogenesis, while follicle-stimulating hormone (FSH) signaling is indispensable for the proliferation, differentiation, and proper functioning of Sertoli and germ cells. This review covers the research on how androgens, FSH, and other hormones support processes crucial for male fertility in the testis and reproductive tract. These hormones are regulated by the hypothalamic-pituitary-gonad (HPG) axis, which is either quiescent or activated at different stages of the life course, and the regulation of the axis is crucial for the development and normal function of the male reproductive system. Hormonal imbalances, whether due to genetic predispositions or environmental influences, leading to hypogonadism or hypergonadism, can precipitate reproductive disorders. Investigating the regulatory network and molecular mechanisms involved in testicular development and spermatogenesis is instrumental in developing new therapeutic methods, drugs, and male hormonal contraceptives.
Collapse
Affiliation(s)
- Lu Li
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
| | - Wanqing Lin
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
| | - Zhaoyang Wang
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
| | - Rufei Huang
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
| | - Huan Xia
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
| | - Ziyi Li
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
| | - Jingxian Deng
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
| | - Tao Ye
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
| | - Yadong Huang
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
- Guangdong Province Key Laboratory of Bioengineering Medicine, Guangzhou 510632, China
- National Engineering Research Center of Genetic Medicine, Guangzhou 510632, China
| | - Yan Yang
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (W.L.); (Z.W.); (R.H.); (H.X.); (Z.L.); (J.D.); (T.Y.)
- Guangdong Province Key Laboratory of Bioengineering Medicine, Guangzhou 510632, China
- National Engineering Research Center of Genetic Medicine, Guangzhou 510632, China
| |
Collapse
|
4
|
Sponchiado M, Fagan A, Mata L, Bonilla AL, Trevizan-Baú P, Prabhakaran S, Reznikov LR. Sex-dependent regulation of mucin gene transcription and airway secretion and mechanics following intra-airway IL-13 in mice with conditional loss of club cell Creb1. Front Physiol 2024; 15:1392443. [PMID: 38711951 PMCID: PMC11070562 DOI: 10.3389/fphys.2024.1392443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 04/01/2024] [Indexed: 05/08/2024] Open
Abstract
Introduction: Interleukin 13 (IL-13) is an important effector molecule in allergic asthma. IL-13-mediated mucin hypersecretion requires conversion of secretoglobin-positive club cells into goblet cells through suppression of forkhead box A2 (FOXA2) and induction of SAM pointed domain containing ETS transcription factor (SPDEF). IL-13-mediated mucin hypersecretion may also include modulation of purinergic and muscarinic receptors that control basal and stimulated mucin secretion. We recently found that the transcription factor cAMP response element-binding protein (Creb1) inhibits FOXA2 and modulates mucus secretion in mice. Methods: We tested the hypothesis that loss of club cell Creb1 mitigates the pro-mucin effects of IL-13. We challenged male and female mice with conditional loss of club cell Creb1 and wild type littermates with intra-airway IL-13 or vehicle. We also studied human "club cell-like" NCI-H322 cells. Results: Loss of club cell Creb1 augmented IL-13-mediated increases in mRNA for the gel-forming mucins Muc5ac and Muc5b and prevented IL-13-mediated decreases in muscarinic 3 receptor (M3R) mRNA in male airways. In female airways, loss of club cell Creb1 reduced M3R mRNA and significantly blunted IL-13-mediated increases in purinergic receptor P2Y2 (P2ry2) mRNA but did not impact Muc5ac and Muc5b mRNA. Despite changes in mucins and secretion machinery, goblet cell density following cholinergic stimulation was not impacted by loss of club cell Creb1 in either sex. IL-13 treatment decreased basal airway resistance across sexes in mice with loss of club cell Creb1, whereas loss of club cell Creb1 augmented IL-13-mediated increases in airway elastance in response to methacholine. NCI-H322 cells displayed IL-13 signaling components, including IL-13Rα1 and IL-4Rα. Pharmacologic inhibition of CREB reduced IL-13Rα1 mRNA, whereas recombinant CREB decreased IL-4Rα mRNA. Application of IL-13 to NCI-H322 cells increased concentrations of cAMP in a delayed manner, thus linking IL-13 signaling to CREB signaling. Conclusion: These data highlight sex-specific regulation of club cell Creb1 on IL-13-mediated mucin hypersecretion and airway mechanics.
Collapse
Affiliation(s)
- Mariana Sponchiado
- Department of Physiological Sciences, University of Florida, Gainesville, FL, United States
| | - Amy Fagan
- Department of Physiological Sciences, University of Florida, Gainesville, FL, United States
| | - Luz Mata
- Department of Physiological Sciences, University of Florida, Gainesville, FL, United States
| | - Angelina L. Bonilla
- Department of Physiological Sciences, University of Florida, Gainesville, FL, United States
| | - Pedro Trevizan-Baú
- Department of Physiological Sciences, University of Florida, Gainesville, FL, United States
| | - Sreekala Prabhakaran
- Department of Pediatrics Pediatric Pulmonary Division, University of Florida, Gainesville, FL, United States
| | - Leah R. Reznikov
- Department of Physiological Sciences, University of Florida, Gainesville, FL, United States
| |
Collapse
|
5
|
Raoofi A, Gholami O, Mokhtari H, Bagheri F, Rustamzadeh A, Nasiry D, Ghaemi A. Caffeine attenuates spermatogenic disorders in mice with induced chronic scrotal hyperthermia. Clin Exp Reprod Med 2024; 51:28-41. [PMID: 38433013 PMCID: PMC10914498 DOI: 10.5653/cerm.2023.06142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 07/20/2023] [Accepted: 09/19/2023] [Indexed: 03/05/2024] Open
Abstract
OBJECTIVE Chronic scrotal hyperthermia (SHT) can lead to serious disorders of the male reproductive system, with oxidative stress playing a key role in the onset of these dysfunctions. Thus, we evaluated the impact of caffeine, a potent antioxidant, on cellular and tissue disorders in mice with chronic SHT. METHODS In this experimental study, 56 adult male NMRI mice were allocated into seven equal groups. Apart from the non-treated control group, all were exposed to heat stress. Two groups, termed "preventive" and "curative," were orally administered caffeine. The preventive mice began receiving caffeine immediately prior to heat exposure, while for the curative group, a caffeine regimen was initiated 15 consecutive days following cessation of heat exposure. Each treated group was subdivided based on pairing with a positive control (Pre/curative [Cur]+PC) or a vehicle (Pre/Cur+vehicle). Upon conclusion of the study, we assessed sperm characteristics, testosterone levels, stereological parameters, apoptosis, antioxidant and oxidant levels, and molecular markers. RESULTS Sperm parameters, testosterone levels, stereological parameters, biochemical factors (excluding malondialdehyde [MDA]), and c-kit gene expression were significantly elevated in the preventive and curative groups, especially the former, relative to the other groups. Conversely, expression levels of the heat shock protein 72 (HSP72) and nuclear factor kappa beta (NF-κβ) genes, MDA levels, and apoptotic cell density were markedly lower in both caffeine-treated groups relative to the other groups, with more pronounced differences observed in the preventive group. CONCLUSION Overall, caffeine attenuated cellular and molecular abnormalities induced by heat stress in the testis, particularly in the mice treated under the preventive condition.
Collapse
Affiliation(s)
- Amir Raoofi
- Cellular and Molecular Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Omid Gholami
- Cellular and Molecular Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Hossein Mokhtari
- Department of Paramedicine, Amol School of Paramedical Sciences, Mazandaran University of Medical Sciences, Sar, Iran
| | - Fatemeh Bagheri
- Student Research Committee, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Auob Rustamzadeh
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Davood Nasiry
- Department of Paramedicine, Amol School of Paramedical Sciences, Mazandaran University of Medical Sciences, Sar, Iran
| | - Alireza Ghaemi
- Department of Basic Sciences and Nutrition, Health Sciences Research Center, Faculty of Public Health, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
6
|
Matsuyama S, DeFalco T. Steroid hormone signaling: multifaceted support of testicular function. Front Cell Dev Biol 2024; 11:1339385. [PMID: 38250327 PMCID: PMC10796553 DOI: 10.3389/fcell.2023.1339385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 12/15/2023] [Indexed: 01/23/2024] Open
Abstract
Embryonic development and adult physiology are dependent on the action of steroid hormones. In particular, the reproductive system is reliant on hormonal signaling to promote gonadal function and to ensure fertility. Here we will describe hormone receptor functions and their impacts on testicular function, focusing on a specific group of essential hormones: androgens, estrogens, progesterone, cortisol, and aldosterone. In addition to focusing on hormone receptor function and localization within the testis, we will highlight the effects of altered receptor signaling, including the consequences of reduced and excess signaling activity. These hormones act through various cellular pathways and receptor types, emphasizing the need for a multifaceted research approach to understand their critical roles in testicular function. Hormones exhibit intricate interactions with each other, as evidenced, for example, by the antagonistic effects of progesterone on mineralocorticoid receptors and cortisol's impact on androgens. In light of research findings in the field demonstrating an intricate interplay between hormones, a systems biology approach is crucial for a nuanced understanding of this complex hormonal network. This review can serve as a resource for further investigation into hormonal support of male reproductive health.
Collapse
Affiliation(s)
- Satoko Matsuyama
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Tony DeFalco
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
7
|
Rey RA. Steroid receptors in the testis: implications in the physiology of prenatal and postnatal development and translation to clinical application. Histol Histopathol 2023; 38:373-389. [PMID: 36218320 DOI: 10.14670/hh-18-533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/01/2023]
Abstract
The testes are the main source of sex steroids in the male, especially androgens and to a lesser extent estrogens. In target cells, steroid hormones typically signal after binding to intracellular receptors, which act as transcription factors. Androgens and estrogens have ubiquitous functions in peripheral organs, but also have paracrine actions within the gonads where they are far more concentrated. The levels of steroid production by the testes vary throughout fetal and postnatal development: they are high in intrauterine life and in the first months after birth, then they decline and are almost undetectable in childhood and increase again during puberty to attain adult levels. The expression of the androgen and estrogen receptors also depict specific ontogenies in the various testicular cell types. The combination of intratesticular steroid concentration with the pattern of expression of the steroid hormone receptors defines androgen and estrogen action on Sertoli, germ and Leydig cells. Here, we review the ontogeny of expression of the androgen and estrogen receptors in the testis, its impact on testicular physiology during prenatal and postnatal development, as well as its implication on the pathophysiology of different disorders affecting gonadal function throughout life.
Collapse
Affiliation(s)
- Rodolfo A Rey
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina.
- Unidad de Medicina Traslacional, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
- Departamento de Biología Celular, Histología, Embriología y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
8
|
Talibova G, Bilmez Y, Ozturk S. Increased double-strand breaks in aged mouse male germ cells may result from changed expression of the genes essential for homologous recombination or nonhomologous end joining repair. Histochem Cell Biol 2023; 159:127-147. [PMID: 36241856 DOI: 10.1007/s00418-022-02157-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2022] [Indexed: 11/26/2022]
Abstract
DNA double-strand breaks (DSBs) are commonly appearing deleterious DNA damages, which progressively increase in male germ cells during biological aging. There are two main pathways for repairing DSBs: homologous recombination (HR) and classical nonhomologous end joining (cNHEJ). Knockout and functional studies revealed that, while RAD51 and RPA70 proteins are indispensable for HR-based repair, KU80 and XRCC4 are the key proteins in cNHEJ repair. As is known, γH2AX contributes to these pathways through recruiting repair-related proteins to damaged site. The underlying reasons of increased DSBs in male germ cells during aging are not fully addressed yet. In this study, we aimed to analyze the spatiotemporal expression of the Rad51, Rpa70, Ku80, and Xrcc4 genes in the postnatal mouse testes, classified into young, prepubertal, pubertal, postpubertal, and aged groups according to their reproductive features and histological structures. We found that expression of these genes significantly decreased in the aged group compared with the other groups (P < 0.05). γH2AX staining showed that DSB levels in the germ cells from spermatogonia to elongated spermatids as well as in the Sertoli cells remarkably increased in the aged group (P < 0.05). The RAD51, RPA70, KU80, and XRCC4 protein levels exhibited predominant changes in the germ and Sertoli cells among groups (P < 0.05). These findings suggest that altered expression of the Rad51, Rpa70, Ku80, and Xrcc4 genes in the germ and Sertoli cells may be associated with increasing DSBs during biological aging, which might result in fertility loss.
Collapse
Affiliation(s)
- Gunel Talibova
- Department of Histology and Embryology, Akdeniz University School of Medicine, Campus, 07070, Antalya, Turkey
| | - Yesim Bilmez
- Department of Histology and Embryology, Akdeniz University School of Medicine, Campus, 07070, Antalya, Turkey
| | - Saffet Ozturk
- Department of Histology and Embryology, Akdeniz University School of Medicine, Campus, 07070, Antalya, Turkey.
| |
Collapse
|
9
|
Aydos OS, Yukselten Y, Ozkan T, Ozkavukcu S, Tuten Erdogan M, Sunguroglu A, Aydos K. Co-Culture of Cryopreserved Healthy Sertoli Cells with Testicular Tissue of Non-Obstructive Azoospermia (NOA) Patients in Culture Media Containing Follicle-Stimulating Hormone (FSH)/Testosterone Has No Advantage in Germ Cell Maturation. J Clin Med 2023; 12:jcm12031073. [PMID: 36769720 PMCID: PMC9917953 DOI: 10.3390/jcm12031073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/11/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023] Open
Abstract
Different cell culture conditions and techniques have been used to mature spermatogenic cells to increase the success of in vitro fertilization. Sertoli cells (SCs) are essential in maintaining spermatogenesis and FSH stimulation exerts its effect through direct or indirect actions on SCs. The effectiveness of FSH and testosterone added to the co-culture has been demonstrated in other studies to provide microenvironment conditions of the testicular niche and to contribute to the maturation and meiotic progression of spermatogonial stem cells (SSCs). In the present study, we investigated whether co-culture of healthy SCs with the patient's testicular tissue in the medium supplemented with FSH/testosterone provides an advantage in the differentiation and maturation of germ cells in NOA cases (N = 34). In men with obstructive azoospermia (N = 12), healthy SCs from testicular biopsies were identified and purified, then cryopreserved. The characterization of healthy SCs was done by flow cytometry (FC) and immunohistochemistry using antibodies specific for GATA4 and vimentin. FITC-conjugated annexin V/PI staining and the MTT assay were performed to compare the viability and proliferation of SCs before and after freezing. In annexin V staining, no difference was found in percentages of live and apoptotic SCs, and MTT showed that cryopreservation did not inhibit SC proliferation compared to the pre-freezing state. Then, tissue samples from NOA patients were processed in two separate environments containing FSH/testosterone and FSH/testosterone plus co-culture with thawed healthy SCs for 7 days. FC was used to measure 7th-day levels of specific markers expressed in spermatogonia (VASA), meiotic cells (CREM), and post-meiotic cells (protamine-2 and acrosin). VASA and acrosin basal levels were found to be lower in infertile patients compared to the OA group (8.2% vs. 30.6% and 12.8% vs. 30.5%, respectively; p < 0.05). Compared to pre-treatment measurements, on the 7th day in the FSH/testosterone environment, CREM levels increased by 58.8% and acrosin levels increased by 195.5% (p < 0.05). Similarly, in medium co-culture with healthy SCs, by day 7, CREM and acrosin levels increased to 92.2% and 204.8%, respectively (p < 0.05). Although VASA and protamine levels increased in both groups, they did not reach a significant level. No significant difference was found between the day 7 increase rates of CREM, VASA, acrosin and protamine-2 in either FSH/testosterone-containing medium or in medium additionally co-cultured with healthy SCs (58.8% vs. 92.2%, 120.6% vs. 79.4%, 195.5% vs. 204.8%, and 232.3% vs. 198.4%, respectively; p > 0.05). Our results suggest that the presence of the patient's own SCs for maturation of germ cells in the culture medium supplemented with FSH and testosterone is sufficient, and co-culture with healthy SCs does not have an additional advantage. In addition, the freezing-thawing process would not impair the viability and proliferation of SCs.
Collapse
Affiliation(s)
- O. Sena Aydos
- Department of Medical Biology, School of Medicine, Ankara University, Ankara 06230, Turkey
- Correspondence: (O.S.A.); (Y.Y.); Tel.: +90-3125958050 (O.S.A.)
| | - Yunus Yukselten
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, CT 06520, USA
- Correspondence: (O.S.A.); (Y.Y.); Tel.: +90-3125958050 (O.S.A.)
| | - Tulin Ozkan
- Department of Medical Biology, School of Medicine, Ankara University, Ankara 06230, Turkey
| | - Sinan Ozkavukcu
- Center for Assisted Reproduction, School of Medicine, Ankara University, Ankara 06230, Turkey
- Postgraduate Medicine, School of Medicine, University of Dundee, Dundee DD1 4HN, UK
| | - Meltem Tuten Erdogan
- Department of Medical Biology, School of Medicine, Ankara University, Ankara 06230, Turkey
| | - Asuman Sunguroglu
- Department of Medical Biology, School of Medicine, Ankara University, Ankara 06230, Turkey
| | - Kaan Aydos
- Department of Urology, School of Medicine, Ankara University, Ankara 06230, Turkey
| |
Collapse
|
10
|
Male Sex Hormones, Metabolic Syndrome, and Aquaporins: A Triad of Players in Male (in)Fertility. Int J Mol Sci 2023; 24:ijms24031960. [PMID: 36768282 PMCID: PMC9915845 DOI: 10.3390/ijms24031960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/12/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023] Open
Abstract
Infertility is becoming a chronic and emerging problem in the world. There is a resistant stigma that this health condition is mostly due to the female, although the literature supports that the responsibility for the onset of infertility is equally shared between both sexes in more or less equal proportions. Nevertheless, male sex hormones, particularly testosterone (T), are key players in male-related infertility. Indeed, hypogonadism, which is also characterized by changes in T levels, is one of the most common causes of male infertility and its incidence has been interconnected to the increased prevalence of metabolic diseases. Recent data also highlight the role of aquaporin (AQP)-mediated water and solute diffusion and the metabolic homeostasis in testicular cells suggesting a strong correlation between AQPs function, metabolism of testicular cells, and infertility. Indeed, recent studies showed that both metabolic and sexual hormone concentrations can change the expression pattern and function of AQPs. Herein, we review up-to-date information on the involvement of AQP-mediated function and permeability in men with metabolic syndrome and testosterone deficit, highlighting the putative mechanisms that show an interaction between sex hormones, AQPs, and metabolic syndrome that may contribute to male infertility.
Collapse
|
11
|
Deng YL, Yang P, Wang YX, Liu C, Luo Q, Shi T, Zeng JY, Lu TT, Chen PP, Miao Y, Zhang M, Cui FP, Lu WQ, Zeng Q. Urinary concentrations of polycyclic aromatic hydrocarbon and phthalate metabolite mixtures in relation to semen quality among men attending an infertility clinic. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:81749-81759. [PMID: 35737263 DOI: 10.1007/s11356-022-21525-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 06/13/2022] [Indexed: 06/15/2023]
Abstract
Previous studies have reported that exposure to phthalates and polycyclic aromatic hydrocarbons (PAHs) is individually associated with altered semen quality, but no human studies have evaluated their joint effects of exposure mixtures, a more real-world scenario. We aimed to explore urinary metabolite mixtures of phthalates and PAHs in associations with semen quality. Repeated spot-urine samples gathered from 695 men attending a fertility clinic were analyzed for urinary metabolites of eight phthalates and ten monohydroxylated-PAHs (OH-PAHs). Principal component analysis (PCA)-multivariable linear regression (MLR) model, quantile g-computation (qg-comp), and Bayesian kernel machine regression (BKMR) were applied to estimate the associations of urinary mixtures of phthalate and OH-PAH metabolites with semen quality. The overall effects of urinary mixtures of phthalate and PAH metabolites on semen quality were not statistically significant. However, hydroxynaphthalene (OHNa) factor identified from PCA was monotonically associated with decreased total sperm count and sperm concentration, whereas di(2-ethylhexyl) phthalate (DEHP) factor was non-monotonically related to increased progressive sperm motility and total sperm motility. Qg-comp and BKMR models confirmed these findings and identified 2-OHNa and 2-OHFlu as the primary negative contributors, whereas MEOHP and MEHP as the primary positive contributors. Our findings suggest that exposure to mixtures of naphthalene and DEHP is associated with altered semen quality. The finding is warranted to confirm in further well-designed epidemiological studies.
Collapse
Affiliation(s)
- Yan-Ling Deng
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Pan Yang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
- Department of Occupational and Environmental Health, School of Medicine, Jinan University, Guangzhou, Guangdong, People's Republic of China
| | - Yi-Xin Wang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Chong Liu
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Qiong Luo
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Tian Shi
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Jia-Yue Zeng
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Ting-Ting Lu
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Pan-Pan Chen
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Yu Miao
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Min Zhang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Fei-Peng Cui
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Wen-Qing Lu
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Qiang Zeng
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China.
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China.
| |
Collapse
|
12
|
Witherspoon L, Flannigan R. It puts the T's in fertility: testosterone and spermatogenesis. Int J Impot Res 2022; 34:669-672. [PMID: 35105947 DOI: 10.1038/s41443-022-00531-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 11/21/2021] [Accepted: 01/17/2022] [Indexed: 11/09/2022]
Affiliation(s)
- Luke Witherspoon
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
- Department of Urology, The Ottawa Hospital, Ottawa, ON, Canada
| | - Ryan Flannigan
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada.
- Department of Urology, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
13
|
Androgen receptor suppresses β-adrenoceptor-mediated CREB activation and thermogenesis in brown adipose tissue of male mice. J Biol Chem 2022; 298:102619. [PMID: 36272644 PMCID: PMC9700029 DOI: 10.1016/j.jbc.2022.102619] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 11/07/2022] Open
Abstract
Thermoregulation is a process by which core body temperature is maintained in mammals. Males typically have a lower body temperature than females. However, the effects of androgens, which show higher levels in males, on adrenergic receptor-mediated thermogenesis remain unclear. Here, we demonstrate that androgen-androgen receptor (AR) signaling suppresses the β-adrenergic agonist-induced rise of core body temperature using castrated and AR knockout (ARKO) male mice. Furthermore, in vitro mechanistic studies show that activated AR inhibits cAMP response element (CRE)-mediated transcription by suppressing cAMP response element-binding protein (CREB) phosphorylation. The elevation of body temperature induced by the β-adrenergic agonist CL316243 was higher in ARKO and castrated mice than in the control mice. Similarly, CL316243 induced a greater increase in Uncoupling protein 1 (Ucp1) expression and CREB phosphorylation in the brown adipose tissue of ARKO mice than in that of controls. We determined that activation of AR by dihydrotestosterone suppressed β3-agonist- or forskolin-induced CRE-mediated transcription, which was prevented by AR antagonist. AR activation also suppressed CREB phosphorylation induced by forskolin. Moreover, we found AR nuclear localization, but not transcriptional activity, was necessary for the suppression of CRE-mediated transcription. Finally, modified mammalian two-hybrid and immunoprecipitation analyses suggest nuclear AR and CREB form a protein complex both in the presence and absence of dihydrotestosterone and forskolin. These results suggest androgen-AR signaling suppresses β-adrenoceptor-induced UCP1-mediated brown adipose tissue thermogenesis by suppressing CREB phosphorylation, presumably owing to a protein complex with AR and CREB. This mechanism explains sexual differences in body temperature, at least partially.
Collapse
|
14
|
Saleki K, Banazadeh M, Saghazadeh A, Rezaei N. Aging, testosterone, and neuroplasticity: friend or foe? Rev Neurosci 2022; 34:247-273. [PMID: 36017670 DOI: 10.1515/revneuro-2022-0033] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 07/03/2022] [Indexed: 11/15/2022]
Abstract
Neuroplasticity or neural plasticity implicates the adaptive potential of the brain in response to extrinsic and intrinsic stimuli. The concept has been utilized in different contexts such as injury and neurological disease. Neuroplasticity mechanisms have been classified into neuroregenerative and function-restoring processes. In the context of injury, neuroplasticity has been defined in three post-injury epochs. Testosterone plays a key yet double-edged role in the regulation of several neuroplasticity alterations. Research has shown that testosterone levels are affected by numerous factors such as age, stress, surgical procedures on gonads, and pharmacological treatments. There is an ongoing debate for testosterone replacement therapy (TRT) in aging men; however, TRT is more useful in young individuals with testosterone deficit and more specific subgroups with cognitive dysfunction. Therefore, it is important to pay early attention to testosterone profile and precisely uncover its harms and benefits. In the present review, we discuss the influence of environmental factors, aging, and gender on testosterone-associated alterations in neuroplasticity, as well as the two-sided actions of testosterone in the nervous system. Finally, we provide practical insights for further study of pharmacological treatments for hormonal disorders focusing on restoring neuroplasticity.
Collapse
Affiliation(s)
- Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, 47176 47745 Babol, Iran.,USERN Office, Babol University of Medical Sciences, 47176 47745 Babol, Iran.,Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), 14197 33151 Tehran, Iran
| | - Mohammad Banazadeh
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), 14197 33151 Tehran, Iran.,Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, 76169 13555 Kerman, Iran
| | - Amene Saghazadeh
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), 14197 33151 Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, 14197 33151 Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, 14197 33151 Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, 14176 13151 Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), 14197 33151 Tehran, Iran
| |
Collapse
|
15
|
Real FM, Lao-Pérez M, Burgos M, Mundlos S, Lupiáñez DG, Jiménez R, Barrionuevo FJ. Cell adhesion and immune response, two main functions altered in the transcriptome of seasonally regressed testes of two mammalian species. JOURNAL OF EXPERIMENTAL ZOOLOGY. PART B, MOLECULAR AND DEVELOPMENTAL EVOLUTION 2022; 340:231-244. [PMID: 35535962 DOI: 10.1002/jez.b.23142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 03/22/2022] [Accepted: 04/09/2022] [Indexed: 12/13/2022]
Abstract
In species with seasonal breeding, male specimens undergo substantial testicular regression during the nonbreeding period of the year. However, the molecular mechanisms that control this biological process are largely unknown. Here, we report a transcriptomic analysis on the Iberian mole, Talpa occidentalis, in which the desquamation of live, nonapoptotic germ cells is the major cellular event responsible for testis regression. By comparing testes at different reproductive states (active, regressing, and inactive), we demonstrate that the molecular pathways controlling the cell adhesion function in the seminiferous epithelium, such as the MAPK, ERK, and TGF-β signaling, are altered during the regression process. In addition, inactive testes display a global upregulation of genes associated with immune response, indicating a selective loss of the "immune privilege" that normally operates in sexually active testes. Interspecies comparative analyses using analogous data from the Mediterranean pine vole, a rodent species where testis regression is controlled by halting meiosis entry, revealed a common gene expression signature in the regressed testes of these two evolutionary distant species. Our study advances in the knowledge of the molecular mechanisms associated to gonadal seasonal breeding, highlighting the existence of a conserved transcriptional program of testis involution across mammalian clades.
Collapse
Affiliation(s)
- Francisca M Real
- Departamento de Genética e Instituto de Biotecnología, Lab. 127, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain.,RG Development & Disease, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Miguel Lao-Pérez
- Departamento de Genética e Instituto de Biotecnología, Lab. 127, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - Miguel Burgos
- Departamento de Genética e Instituto de Biotecnología, Lab. 127, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - Stefan Mundlos
- RG Development & Disease, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Darío G Lupiáñez
- Epigenetics and Sex Development Group, Max-Delbrück Center for Molecular Medicine, Berlin Institute for Medical Systems Biology, Berlin, Germany
| | - Rafael Jiménez
- Departamento de Genética e Instituto de Biotecnología, Lab. 127, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - Francisco J Barrionuevo
- Departamento de Genética e Instituto de Biotecnología, Lab. 127, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| |
Collapse
|
16
|
Abstract
Male infertility contributes to 50% of all cases of infertility. The main cause is low quality and quantity of sperm. In humans, spermatogenesis starts at the beginning of puberty and lasts lifelong. It is under the control of FSH and testicular androgens, and mainly testosterone (T), and therefore requires a normal gonadotroph axis, intratesticular T production by Leydig cells and functional androgen receptors (ARs) within testicular Sertoli cells. Various clinical cases illustrate the roles of T in human spermatogenesis. Men with complete congenital hypogonadotropic hypogonadism (HH) are usually azoospermic. Treatment by exogenous testosterone injection and FSH is not able to produce sperm. However, combined treatment with FSH and hCG is effective. This example shows that intratesticular T plays a major role in spermatogenesis. Furthermore, testicular histology of men with LH receptor mutations shows Leydig cell hypoplasia/agenesis/dysplasia with conserved Sertoli cell count. The sperm count is reduced, as in males with partial inactivating mutation of the androgen receptor. Some protocols of hormonal male contraception or exogenous androgen abuse induce negative feedback in the hypothalamic pituitary axis, decreasing FSH, LH and T levels and inducing sperm defects and testicular atrophy. The time to recovery after cessation of drug abuse is around 14 months for sperm output and 38 months for sperm motility. In summary, abnormal androgen production and/or AR signaling impairs spermatogenesis in humans. The minimal level of intratesticular T for normal sperm production is a matter of debate. Interestingly, some animal models showed that completely T-independent spermatogenesis is possible, potentially through strong FSH activation. Finally, recent data suggest important roles of prenatal life and minipuberty in adult spermatogenesis.
Collapse
|
17
|
Abstract
Hypogonadism can be present in up to 40% in men who present with couple infertility. Testosterone is the major androgen regulating-spermatogenesis in men; as a result, men with either primary or secondary hypogonadism may be subfertile because of impaired spermatogenesis. The clinical impact of hypogonadism on fertility potential depends on the timing of its onset (fetal, prepubertal, or postpubertal) and effect on semen parameters. Treatment pathways and success rates differ according to the cause of hypogonadism and the time of its onset. When medical therapy fails to induce sufficient sperm, assisted reproductive technologies are considered.
Collapse
Affiliation(s)
- Nikoleta Papanikolaou
- Section of Investigative Medicine Imperial College London, Hammersmith Hospital, 6th Floor, Commonwealth Building, 150 Du Cane Road, London W12 0NN, UK
| | - Rong Luo
- Section of Investigative Medicine Imperial College London, Hammersmith Hospital, 6th Floor, Commonwealth Building, 150 Du Cane Road, London W12 0NN, UK
| | - Channa N Jayasena
- Section of Investigative Medicine Imperial College London, Hammersmith Hospital, 6th Floor, Commonwealth Building, 150 Du Cane Road, London W12 0NN, UK.
| |
Collapse
|
18
|
Wang JM, Li ZF, Yang WX. What Does Androgen Receptor Signaling Pathway in Sertoli Cells During Normal Spermatogenesis Tell Us? Front Endocrinol (Lausanne) 2022; 13:838858. [PMID: 35282467 PMCID: PMC8908322 DOI: 10.3389/fendo.2022.838858] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 02/01/2022] [Indexed: 01/18/2023] Open
Abstract
Androgen receptor signaling pathway is necessary to complete spermatogenesis in testes. Difference between androgen binding location in Sertoli cell classifies androgen receptor signaling pathway into classical signaling pathway and non-classical signaling pathway. As the only somatic cell type in seminiferous tubule, Sertoli cells are under androgen receptor signaling pathway regulation via androgen receptor located in cytoplasm and plasma membrane. Androgen receptor signaling pathway is able to regulate biological processes in Sertoli cells as well as germ cells surrounded between Sertoli cells. Our review will summarize the major discoveries of androgen receptor signaling pathway in Sertoli cells and the paracrine action on germ cells. Androgen receptor signaling pathway regulates Sertoli cell proliferation and maturation, as well as maintain the integrity of blood-testis barrier formed between Sertoli cells. Also, Spermatogonia stem cells achieve a balance between self-renewal and differentiation under androgen receptor signaling regulation. Meiotic and post-meiotic processes including Sertoli cell - Spermatid attachment and Spermatid development are guaranteed by androgen receptor signaling until the final sperm release. This review also includes one disease related to androgen receptor signaling dysfunction named as androgen insensitivity syndrome. As a step further ahead, this review may be conducive to develop therapies which can cure impaired androgen receptor signaling in Sertoli cells.
Collapse
|
19
|
Kuwahara N, Nicholson K, Isaacs L, MacLusky NJ. Androgen Effects on Neural Plasticity. ANDROGENS: CLINICAL RESEARCH AND THERAPEUTICS 2021; 2:216-230. [PMID: 35024693 PMCID: PMC8744448 DOI: 10.1089/andro.2021.0022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 10/24/2021] [Indexed: 12/20/2022]
Abstract
Androgens are synthesized in the brain, gonads, and adrenal glands, in both sexes, exerting physiologically important effects on the structure and function of the central nervous system. These effects may contribute to the incidence and progression of neurological disorders such as autism spectrum disorder, schizophrenia, and Alzheimer's disease, which occur at different rates in males and females. This review briefly summarizes the current state of knowledge with respect to the neuroplastic effects of androgens, with particular emphasis on the hippocampus, which has been the focus of much of the research in this field.
Collapse
Affiliation(s)
- Nariko Kuwahara
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Kate Nicholson
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Lauren Isaacs
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Neil J. MacLusky
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| |
Collapse
|
20
|
Xiao L, Sun W, Su Y, Lu N, He Y, Sheng X, Qi X, Xing K, Guo Y, Chang D, Wang X, Zhao J, Ni H. Dihydrotestosterone regulation of cyclooxygenase-2 expression in bovine endometrial epithelium cells by androgen receptor mediated EGFR/PI3K/Akt pathway. J Steroid Biochem Mol Biol 2021; 214:106001. [PMID: 34547381 DOI: 10.1016/j.jsbmb.2021.106001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 09/07/2021] [Accepted: 09/15/2021] [Indexed: 11/29/2022]
Abstract
Uterine prostaglandins F2α (PGF2α) is essential for implantation, initiation of luteolysis and delivery. Previous studies have demonstrated that the expression of Cyclooxygenase-2 (COX-2), an enzyme limiting PGF2α rate, is regulated by steroid hormones, and also dihydrotestosterone (DHT) may be involved in regulating COX-2 expression both positively and negatively. However, it remains unclear how whether DHT regulates COX-2 expression and consequent PGF2α release in bovine endometrial epithelial cells (EECs). In this study, we evaluated the localization of the two isoforms of DHT synthetase 5α-reductase (5α-red1 and 5α-red2) and androgen receptor (AR) in bovine endometria by immunohistochemistry, and investigated 5α-red1, 5α-red2, AR, and DHT levels at the different stages of endometria (follicle, early-, mid-, and late-pregnancy phases). The results showed that 5α-red1, 5α-red2 and AR all were expressed in endometria, and their expressions and the level of DHT significantly increased in the late-pregnancy phase compared with the mid-pregnancy phase. Moreover, we cultured EECs from the mid-pregnancy phase and the in vitro study showed that DHT dose-dependently increased COX-2 expression and PGF2a release, but AR antagonist (flutamide) inhibited the stimulating effect via DHT. In addition, the DHT-induced COX-2 expression and PGF2α release were subjected to the regulation of both EGFR/PI3K/Akt/NFkB signaling as the inhibitors of EGFR (AG1478) and PI3K/Akt (LY294002) and NFkB (QNZ) attenuated the DHT mediated effect. Taken together, the results demonstrated that DHT-induced COX-2 expression and consequent PGF2α release in bovine EECs were mediated through AR-derived EGFR transactivation and PI3K/Akt cascade leading to NFkB activation.
Collapse
Affiliation(s)
- Longfei Xiao
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - Wanxu Sun
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - Yue Su
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - Ning Lu
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - Yanan He
- Zhangjiagang Agriculture and Rural Affairs Bureau, Jiangsu, China
| | - Xihui Sheng
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - Xiaolong Qi
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - Kai Xing
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - Yong Guo
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - Di Chang
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - Xiangguo Wang
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China.
| | - Junjin Zhao
- National Grazing Headquarter, Beijing, China
| | - Hemin Ni
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China.
| |
Collapse
|
21
|
Wang Z, Wu W, Kim MS, Cai D. GnRH pulse frequency and irregularity play a role in male aging. NATURE AGING 2021; 1:904-918. [PMID: 37118330 DOI: 10.1038/s43587-021-00116-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 08/12/2021] [Indexed: 04/30/2023]
Abstract
Gonadotropin-releasing hormone (GnRH) has a role in hypothalamic control of aging, but the underlying patterns and relationship with downstream reproductive hormones are still unclear. Here we report that hypothalamic GnRH pulse frequency and irregularity increase before GnRH pulse amplitude slowly decreases during aging. GnRH is inhibited by nuclear factor (NF)-κB, and GnRH pulses were controlled by oscillations in the transcriptional activity of NF-κB. Exposure to testosterone under pro-inflammatory conditions stimulated both NF-κB oscillations and GnRH pulses. While castration of middle-aged mice induced short-term anti-aging effects, preventing elevation of luteinizing hormone (LH) levels after castration led to long-term anti-aging effects and lifespan extension, indicating that high-frequency GnRH pulses and high-magnitude LH levels coordinately mediate aging. Reprogramming the endogenous GnRH pulses of middle-aged male mice via an optogenetic approach revealed that increasing GnRH pulses frequency causes LH excess and aging acceleration, while lowering the frequency of and stabilizing GnRH pulses can slow down aging. In conclusion, GnRH pulses are important for aging in male mice.
Collapse
Affiliation(s)
- Zhouguang Wang
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Wenhe Wu
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Min Soo Kim
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Dongsheng Cai
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
22
|
Cooke PS, Walker WH. Male fertility in mice requires classical and nonclassical androgen signaling. Cell Rep 2021; 36:109557. [PMID: 34407397 DOI: 10.1016/j.celrep.2021.109557] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 06/03/2021] [Accepted: 07/27/2021] [Indexed: 11/22/2022] Open
Abstract
Molecular mechanisms by which androgens signal through the androgen receptor (AR) to maintain male fertility are poorly understood. Transgenic mice were produced expressing mutant ARs that can only (1) alter gene transcription through the classical response pathway (AR-C) or (2) activate kinase signaling cascades via the nonclassical pathway (AR-NC). AR-C is sufficient to produce sperm and fertility. Haploid germ cell production, the blood-testis barrier, and spermatid migration are supported by AR-NC. Gene expression essential for chromosome synapsis during meiosis requires AR-C. We identify targets of androgen signaling required for male fertility and provide a mechanistic explanation for meiotic germ cell arrest in the absence of androgen signaling. Prostate differentiation occurs with AR-C alone, but full development requires synergistic nonclassical signaling. Both AR signaling pathways are necessary for normal male reproductive tract development and function, validating our mouse models for studies of AR functions in other target tissues.
Collapse
Affiliation(s)
- Paul S Cooke
- Department of Physiological Sciences, University of Florida, Gainesville, FL 32610, USA
| | - William H Walker
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Magee-Womens Research Institute, Pittsburgh, PA 15213, USA.
| |
Collapse
|
23
|
Nonclassical androgen and estrogen signaling is essential for normal spermatogenesis. Semin Cell Dev Biol 2021; 121:71-81. [PMID: 34119408 DOI: 10.1016/j.semcdb.2021.05.032] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 05/25/2021] [Accepted: 05/31/2021] [Indexed: 11/20/2022]
Abstract
Signaling by androgens through androgen receptor (AR) is essential to complete spermatogenesis in the testis. Similarly, loss of the main estrogen receptor, estrogen receptor 1 (ESR1; also known as ERα), results in male infertility, due in part to indirect deleterious effects on the seminiferous epithelium and spermatogenesis. Effects of steroid hormones are induced primarily through genomic changes induced by hormone-mediated activation of their intracellular receptors and subsequent effects on nuclear gene transcription. However, androgens and estrogens also signal through rapid nonclassical pathways involving actions initiated at the cell membrane. Here we review the data that nonclassical androgen and estrogen signaling pathways support processes essential for male fertility in the testis and reproductive tract. The recent development of transgenic mice lacking nonclassical AR or ESR1 signaling but retaining genomic nuclear signaling has provided a powerful tool to elucidate the function of nonclassical signaling in the overall response to androgens and estrogens. Results from these mice have emphasized that nonclassical signaling is essential for full responses to these hormones, and absence of either nonclassical or classical AR or ESR1 pathways produces abnormalities in spermatogenesis and the male reproductive tract. Although additional work is required to fully understand how classical and nonclassical receptor signaling synergize to produce full steroid hormone responses, here we summarize the known physiological functions of the classical and nonclassical androgen and estrogen signaling pathways in the testis and reproductive tract.
Collapse
|
24
|
Mediterranean Pine Vole, Microtus duodecimcostatus: A Paradigm of an Opportunistic Breeder. Animals (Basel) 2021; 11:ani11061639. [PMID: 34205873 PMCID: PMC8228771 DOI: 10.3390/ani11061639] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 05/23/2021] [Accepted: 05/27/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary In temperate zones of the Earth, some mammalian species reproduce seasonally whereas others do it continuously. Other species are summer breeders in the north and winter breeders in the south. Thus, the reproductive pattern seems not to be a species-specific but a population-specific trait. We investigated the reproduction pattern of the Mediterranean pine vole, Microtus duodecimcostatus, in the area around the city of Granada in Southern Spain, and found that individuals living in wastelands reproduce seasonally whereas those living in close poplar plantations (just 8 km apart) reproduce throughout the year, as did voles captured in wastelands and kept in captivity. These animals represent thus a paradigm of an opportunistic breeder as particular individuals stop breeding or not, depending on the environmental conditions they face at any moment. Sexually inactive male voles undergo complete testis inactivation and their sperm production is halted. The immune system in active testes is depressed, a phenomenon known as “immune privilege” that protect germ cells from autoimmune attack. We studied gene activity in active and inactive testes and our results indicate that such an immune privilege is lost in inactive testes, suggesting an important role for this process during testis regression. Abstract Most mammalian species of the temperate zones of the Earth reproduce seasonally, existing a non-breeding period in which the gonads of both sexes undergo functional regression. It is widely accepted that photoperiod is the principal environmental cue controlling these seasonal changes, although several exceptions have been described in other mammalian species in which breeding depends on cues such as food or water availability. We studied the circannual reproductive cycle in males of the Mediterranean pine vole, Microtus duodecimcostatus, in the Southeastern Iberian Peninsula. Morphological, hormonal, functional, molecular and transcriptomic analyses were performed. As reported for populations of other species from the same geographic area, male voles captured in wastelands underwent seasonal testis regression in summer whereas, surprisingly, those living either in close poplar plantations or in our animal house reproduced throughout the year, showing that it is the microenvironment of a particular vole subpopulation what determines its reproductive status and that these animals are pure opportunistic, photoperiod-independent breeders. In addition, we show that several molecular pathways, including MAPK, are deregulated and that the testicular “immune privilege” is lost in the inactive testes, providing novel mechanisms linking seasonal testosterone reduction and testis regression.
Collapse
|
25
|
Levkova M, Chervenkov T, Hachmeriyan M, Angelova L. CFTR gene variants as a reason for impaired spermatogenesis: a pilot study and a Meta-analysis of published data. HUM FERTIL 2021; 25:728-737. [PMID: 33719834 DOI: 10.1080/14647273.2021.1900608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
There is increasing data that IVS8-5T variand and TG repeats could lead to impaired spermatogenesis. To investigate this we performed Sanger sequencing on 50 Bulgarian men with a sperm count below 5 × 106/mL and 20 normal fertile men. Frequencies of the results were compared among the two groups. A meta-analysis was perfomed by using the data for 6,423 patients and 5,834 control subjects, tested for the IVS8-5T polymorphism. One case subject (2.0%) was homozygote for the 5 T/5T variant whereas two (4.0%) were heterozygotes for the 5 T/7T variant. No 5 T alleles were found in the control group. The genotypes of the two groups showed a statistically significant difference (p = 0.04, α < 0.05). Also, the odds ratio was 3.73, but this was unsignificant (p = 0.38). All control subjects had 11 TG repeats and for the test group: 47 (94.0%) men with 11 TG repeats and three (6.00%) with 10 TG repeats. Fisher's test showed no significant difference (p = 0.55). The meta-analysis showed that IVS8-5T variant was a risk factor for impaired spermatogenesis (OR = 2.84, p < 0.05) and this was more prominent for non-European (OR = 4.50, p < 0.05) compared to European (OR = 1.28, p < 0.05) men. The IVS8 - 5 T variant could be associated with disorders of sperm production.
Collapse
Affiliation(s)
- Mariya Levkova
- Department of Medical Genetics, Medical University Varna, Varna, Bulgaria.,Laboratory of Medical Genetics, St. Marina Hospital, Varna, Bulgaria
| | - Trifon Chervenkov
- Department of Medical Genetics, Medical University Varna, Varna, Bulgaria.,Laboratory of Clinical Immunology, St. Marina Hospital, Varna, Bulgaria
| | - Mari Hachmeriyan
- Department of Medical Genetics, Medical University Varna, Varna, Bulgaria.,Laboratory of Medical Genetics, St. Marina Hospital, Varna, Bulgaria
| | - Lyudmila Angelova
- Department of Medical Genetics, Medical University Varna, Varna, Bulgaria
| |
Collapse
|
26
|
Abstract
Puberty is characterized by major changes in the anatomy and function of reproductive organs. Androgen activity is low before puberty, but during pubertal development, the testes resume the production of androgens. Major physiological changes occur in the testicular cell compartments in response to the increase in intratesticular testosterone concentrations and androgen receptor expression. Androgen activity also impacts on the internal and external genitalia. In target cells, androgens signal through a classical and a nonclassical pathway. This review addresses the most recent advances in the knowledge of the role of androgen signaling in postnatal male sexual development, with a special emphasis on human puberty.
Collapse
Affiliation(s)
- Rodolfo A Rey
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Histología, Embriología, Biología Celular y Genética, C1121ABG Buenos Aires, Argentina
| |
Collapse
|
27
|
Mahdivand N, Shalizar-Jalali A, Nejati V, Najafi G, Rahmani F. Adaptogenic potential of royal jelly in reproductive system of heat stress-exposed male rats. J Therm Biol 2021; 96:102827. [PMID: 33627267 DOI: 10.1016/j.jtherbio.2020.102827] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 11/23/2020] [Accepted: 12/31/2020] [Indexed: 11/16/2022]
Abstract
Testicular heat stress (HS) can lead to testicular tissue destruction and spermatogenesis disturbances. Royal Jelly (RJ) has been introduced as a potent antioxidant. We investigated the effects of RJ on testicular tissue, oxidative stress and sperm apoptosis in HS-exposed rats. Compared to HS-exposed groups, RJ co-treatment could improve testosterone reduction and histopathological damages. The RJ co-administration decreased MDA level in testicular tissue, while TAC and CAT levels were remarkably increased compared to HS-exposed groups. Moreover, significant higher expression level of Bcl-2 and lower expression levels of P53 and Caspase-3 were seen following RJ co-administration compared to HS-exposed groups. Our data suggest that RJ can effectively ameliorate experimental HS-induced testiculopathies in rats through testicular antioxidant defense system restoration and germ cells apoptosis regulation.
Collapse
Affiliation(s)
- Noushin Mahdivand
- Department of Biology, Faculty of Science, Urmia University, Urmia, Iran
| | - Ali Shalizar-Jalali
- Department of Basic Sciences, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran.
| | - Vahid Nejati
- Department of Biology, Faculty of Science, Urmia University, Urmia, Iran
| | - Gholamreza Najafi
- Department of Basic Sciences, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Fatemeh Rahmani
- Department of Biology, Faculty of Science, Urmia University, Urmia, Iran
| |
Collapse
|
28
|
Walker WH. Androgen Actions in the Testis and the Regulation of Spermatogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1288:175-203. [PMID: 34453737 DOI: 10.1007/978-3-030-77779-1_9] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Testosterone is essential for spermatogenesis and male fertility. In this review, topics related to testosterone control of spermatogenesis are covered including testosterone production and levels in the testis, classical and nonclassical testosterone signaling pathways, cell- and temporal-specific expression of the androgen receptor in the testis and autocrine and paracrine signaling of testis cells in the testis. Also discussed are the contributions of testosterone to testis descent, the blood-testis barrier, control of gonocyte numbers and spermatogonia expansion, completion of meiosis and attachment and release of elongaed spermatids. Testosterone-regulated genes identified in various mouse models of idsrupted Androgen receptor expression are discussed. Finally, examples of synergism and antagonism between androgen and follicle-stimulating hormone signaling pathways are summarized.
Collapse
Affiliation(s)
- William H Walker
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
29
|
Shah W, Khan R, Shah B, Khan A, Dil S, Liu W, Wen J, Jiang X. The Molecular Mechanism of Sex Hormones on Sertoli Cell Development and Proliferation. Front Endocrinol (Lausanne) 2021; 12:648141. [PMID: 34367061 PMCID: PMC8344352 DOI: 10.3389/fendo.2021.648141] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 05/17/2021] [Indexed: 12/30/2022] Open
Abstract
Sustaining and maintaining the intricate process of spermatogenesis is liable upon hormones and growth factors acting through endocrine and paracrine pathways. The Sertoli cells (SCs) are the major somatic cells present in the seminiferous tubules and are considered to be the main regulators of spermatogenesis. As each Sertoli cell supports a specific number of germ cells, thus, the final number of Sertoli cells determines the sperm production capacity. Similarly, sex hormones are also major regulators of spermatogenesis and they can determine the proliferation of Sertoli cells. In the present review, we have critically and comprehensively discussed the role of sex hormones and some other factors that are involved in Sertoli cell proliferation, differentiation and maturation. Furthermore, we have also presented a model of Sertoli cell development based upon the recent advancement in the field of reproduction. Hence, our review article provides a general overview regarding the sex hormonal pathways governing Sertoli cell proliferation and development.
Collapse
Affiliation(s)
| | - Ranjha Khan
- *Correspondence: Xiaohua Jiang, ; Ranjha Khan, ; Jie Wen,
| | | | | | | | | | - Jie Wen
- *Correspondence: Xiaohua Jiang, ; Ranjha Khan, ; Jie Wen,
| | - Xiaohua Jiang
- *Correspondence: Xiaohua Jiang, ; Ranjha Khan, ; Jie Wen,
| |
Collapse
|
30
|
Cooke PS, Mesa AM, Sirohi VK, Levin ER. Role of nuclear and membrane estrogen signaling pathways in the male and female reproductive tract. Differentiation 2020; 118:24-33. [PMID: 33339644 DOI: 10.1016/j.diff.2020.11.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 11/02/2020] [Indexed: 12/13/2022]
Abstract
Estrogen signaling through the main estrogen receptor, estrogen receptor 1 (ESR1; also known as ERα), is essential for normal female and male reproductive function. Historically, studies of estrogen action have focused on the classical genomic pathway. Although this is clearly the major pathway for steroid hormone actions, these hormones also signal through rapid non-classical effects involving cell membrane actions. Reports of rapid effects of estrogens extend for more than half a century, but recent results have expanded understanding of the identity, structure, function and overall importance of membrane receptors in estrogen responses. Key findings in this field were the immunohistochemical detection of ESR1 in cell membranes and demonstration that a portion of newly synthesized ESR1 is routed to the membrane by palmitoylation. These receptors in the membrane can then signal through protein kinases and other mechanisms following ligand binding to alter cell function. Another crucial advance in the field was development of transgenic mice expressing normal amounts of functional nuclear ESR1 (nESR1) but lacking membrane ESR1 (mESR1). Both male and female transgenic mice lacking mESR1 were infertile as adults, and both sexes had extensive reproductive abnormalities. Transgenic mice lacking mESR1 were highly protected from deleterious effects of neonatal estrogen administration, and estrogen effects on the histone methyltransferase Enhancer of Zeste homolog 2 that are mediated through mESR1 could have significant effects on epigenetic imprinting. In summary, signaling through mESR1 is essential for normal male and female reproductive function and fertility, and is a critical enabler of normal estrogen responses in vivo. Although the precise role of mESR1 in estrogen responses remains to be established, future research in this area should clarify its mechanism of action and lead to a better understanding of how mESR1 signaling works with classical genomic signaling through nESR1 to promote full estrogenic responses.
Collapse
Affiliation(s)
- Paul S Cooke
- Department of Physiological Sciences, University of Florida, Gainesville, FL, 32610, USA.
| | - Ana M Mesa
- Department of Physiological Sciences, University of Florida, Gainesville, FL, 32610, USA
| | - Vijay K Sirohi
- Department of Physiological Sciences, University of Florida, Gainesville, FL, 32610, USA
| | - Ellis R Levin
- Division of Endocrinology, Department of Medicine, University of California, Irvine, Irvine, CA, 92697, USA; Department of Veterans Affairs Medical Center, Long Beach, Long Beach, CA, 90822, USA
| |
Collapse
|
31
|
Singh JP, Dagar M, Dagar G, Kumar S, Rawal S, Sharma RD, Tyagi RK, Bagchi G. Activation of GPR56, a novel adhesion GPCR, is necessary for nuclear androgen receptor signaling in prostate cells. PLoS One 2020; 15:e0226056. [PMID: 32881870 PMCID: PMC7470385 DOI: 10.1371/journal.pone.0226056] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 07/08/2020] [Indexed: 12/18/2022] Open
Abstract
The androgen receptor (AR) is activated in patients with castration resistant prostate cancer (CRPC) despite low circulating levels of androgen, suggesting that intracellular signaling pathways and non-androgenic factors may contribute to AR activation. Many G-protein coupled receptors (GPCR) and their ligands are also activated in these cells indicating that they may play a role in development of Prostate Cancer (PCa) and CRPC. Although a cross talk has been suggested between the two pathways, yet, the identity of GPCRs which may play a role in androgen signaling, is not established yet. By using blast analysis of 826 GPCRs, we identified a GPCR, GPCR 205, which exhibited maximum similarity with the ligand binding domain of the AR. We demonstrate that adhesion GPCR 205, also known as GPR56, can be activated by androgens to stimulate the Rho signaling pathway, a pathway that plays an important role in prostate tumor cell metastasis. Testosterone stimulation of GPR56 also activates the cAMP/ Protein kinase A (PKA) pathway, that is necessary for AR signaling. Knocking down the expression of GPR56 using siRNA, disrupts nuclear translocation of AR and transcription of prototypic AR target genes such as PSA. GPR56 expression is higher in all twenty-five prostate tumor patient's samples tested and cells expressing GPR56 exhibit increased proliferation. These findings provide new insights about androgen signaling and identify GPR56 as a possible therapeutic target in advanced prostate cancer patients.
Collapse
MESH Headings
- Aged
- Androgens/metabolism
- Animals
- COS Cells
- Cell Line, Tumor
- Cell Nucleus/metabolism
- Chlorocebus aethiops
- Gene Expression Regulation, Neoplastic
- Gene Knockdown Techniques
- HEK293 Cells
- Humans
- Male
- Middle Aged
- Molecular Docking Simulation
- Prostate/cytology
- Prostate/pathology
- Prostate/surgery
- Prostatectomy
- Prostatic Neoplasms, Castration-Resistant/genetics
- Prostatic Neoplasms, Castration-Resistant/pathology
- Prostatic Neoplasms, Castration-Resistant/surgery
- RNA, Small Interfering/metabolism
- Receptors, Androgen/metabolism
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Signal Transduction/genetics
- Testosterone/metabolism
- Transcription, Genetic
Collapse
Affiliation(s)
- Julie Pratibha Singh
- Amity Institute of Biotechnology (AIB), Amity University Haryana, Manesar, Gurugram, India
| | - Manisha Dagar
- Amity Institute of Biotechnology (AIB), Amity University Haryana, Manesar, Gurugram, India
| | - Gunjan Dagar
- Amity Institute of Biotechnology (AIB), Amity University Haryana, Manesar, Gurugram, India
| | - Sudhir Kumar
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Sudhir Rawal
- Rajiv Gandhi Cancer Institute & Research Centre, Rohini, New Delhi, India
| | - Ravi Datta Sharma
- Amity Institute of Integrative Sciences and Health (AIISH), Amity University Haryana, Manesar, Gurugram, India
| | - Rakesh Kumar Tyagi
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Gargi Bagchi
- Amity Institute of Biotechnology (AIB), Amity University Haryana, Manesar, Gurugram, India
| |
Collapse
|
32
|
Molecular insights into hormone regulation via signaling pathways in Sertoli cells: With discussion on infertility and testicular tumor. Gene 2020; 753:144812. [DOI: 10.1016/j.gene.2020.144812] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 05/17/2020] [Accepted: 05/22/2020] [Indexed: 02/07/2023]
|
33
|
Amiri S, Azadmanesh K, Dehghan Shasaltaneh M, Mayahi V, Naghdi N. The Implication of Androgens in the Presence of Protein Kinase C to Repair Alzheimer’s Disease-Induced Cognitive Dysfunction. IRANIAN BIOMEDICAL JOURNAL 2020; 24:64-80. [PMID: 31677609 PMCID: PMC6984714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 06/10/2019] [Indexed: 03/29/2024]
Abstract
Aging, as a major risk factor of memory deficiency, affects neural signaling pathways in hippocampus. In particular, age-dependent androgens deficiency causes cognitive impairments. Several enzymes like protein kinase C (PKC) are involved in memory deficiency. Indeed, PKC regulatory process mediates α-secretase activation to cleave APP in β-amyloid cascade and tau proteins phosphorylation mechanism. Androgens and cortisol regulate PKC signaling pathways, affecting the modulation of receptor for activated C kinase 1. Mitogen-activated protein kinase/ERK signaling pathway depends on CREB activity in hippocampal neurons and is involved in regulatory processes via PKC and androgens. Therefore, testosterone and PKC contribute in the neuronal apoptosis. The present review summarizes the current status of androgens, PKC, and their influence on cognitive learning. Inconsistencies in experimental investigations related to this fundamental correlation are also discussed, with emphasis on the mentioned contributors as the probable potent candidates for learning and memory improvement.
Collapse
Affiliation(s)
- Sara Amiri
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| | | | | | - Vafa Mayahi
- Department of Microbiology, Islamic Azad University, Karaj, Iran
| | - Nasser Naghdi
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
34
|
Gegenhuber B, Tollkuhn J. Signatures of sex: Sex differences in gene expression in the vertebrate brain. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2020; 9:e348. [PMID: 31106965 PMCID: PMC6864223 DOI: 10.1002/wdev.348] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/10/2019] [Accepted: 04/22/2019] [Indexed: 12/13/2022]
Abstract
Women and men differ in disease prevalence, symptoms, and progression rates for many psychiatric and neurological disorders. As more preclinical studies include both sexes in experimental design, an increasing number of sex differences in physiology and behavior have been reported. In the brain, sex-typical behaviors are thought to result from sex-specific patterns of neural activity in response to the same sensory stimulus or context. These differential firing patterns likely arise as a consequence of underlying anatomic or molecular sex differences. Accordingly, gene expression in the brains of females and males has been extensively investigated, with the goal of identifying biological pathways that specify or modulate sex differences in brain function. However, there is surprisingly little consensus on sex-biased genes across studies and only a handful of robust candidates have been pursued in the follow-up experiments. Furthermore, it is not known how or when sex-biased gene expression originates, as few studies have been performed in the developing brain. Here we integrate molecular genetic and neural circuit perspectives to provide a conceptual framework of how sex differences in gene expression can arise in the brain. We detail mechanisms of gene regulation by steroid hormones, highlight landmark studies in rodents and humans, identify emerging themes, and offer recommendations for future research. This article is categorized under: Nervous System Development > Vertebrates: General Principles Gene Expression and Transcriptional Hierarchies > Regulatory Mechanisms Gene Expression and Transcriptional Hierarchies > Sex Determination.
Collapse
Affiliation(s)
- Bruno Gegenhuber
- Watson School of Biological Sciences, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | | |
Collapse
|
35
|
Ohya S, Ito K, Hatano N, Ohno A, Muraki K, Imaizumi Y. Castration Induces Down-Regulation of A-Type K + Channel in Rat Vas Deferens Smooth Muscle. Int J Mol Sci 2019; 20:ijms20174073. [PMID: 31438481 PMCID: PMC6747096 DOI: 10.3390/ijms20174073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 08/03/2019] [Accepted: 08/19/2019] [Indexed: 01/20/2023] Open
Abstract
A-type K+ channels contribute to regulating the propagation and frequency of action potentials in smooth muscle cells (SMCs). The present study (i) identified the molecular components of A-type K+ channels in rat vas deferens SMs (VDSMs) and (ii) showed the long-term, genomic effects of testosterone on their expression in VDSMs. Transcripts of the A-type K+ channel α subunit, Kv4.3L and its regulatory β subunits, KChIP3, NCS1, and DPP6-S were predominantly expressed in rat VDSMs over the other related subtypes (Kv4.2, KChIP1, KChIP2, KChIP4, and DPP10). A-type K+ current (IA) density in VDSM cells (VDSMCs) was decreased by castration without changes in IA kinetics, and decreased IA density was compensated for by an oral treatment with 17α-methyltestosterone (MET). Correspondingly, in the VDSMs of castrated rats, Kv4.3L and KChIP3 were down-regulated at both the transcript and protein expression levels. Changes in Kv4.3L and KChIP3 expression levels were compensated for by the treatment with MET. These results suggest that testosterone level changes in testosterone disorders and growth processes control the functional expression of A-type K+ channels in VDSMCs.
Collapse
Affiliation(s)
- Susumu Ohya
- Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan.
| | - Katsunori Ito
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmacological Sciences, Nagoya City University, Nagoya 467-8603, Japan
| | - Noriyuki Hatano
- Laboratory of Cellular Pharmacology, Aichi Gakuin University, Nagoya 464-8650, Japan
| | - Akitoshi Ohno
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmacological Sciences, Nagoya City University, Nagoya 467-8603, Japan
| | - Katsuhiko Muraki
- Laboratory of Cellular Pharmacology, Aichi Gakuin University, Nagoya 464-8650, Japan
| | - Yuji Imaizumi
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmacological Sciences, Nagoya City University, Nagoya 467-8603, Japan.
| |
Collapse
|
36
|
Wang Q, Wang HX, Shen JY, Zhang R, Hong JW, Li Z, Chen G, Li MX, Ding Z, Li J, Zhang JP, Zhang MR, Xu LC. The anti-androgenic effects of cypermethrin mediated by non-classical testosterone pathway activation of mitogen-activated protein kinase cascade in mouse Sertoli cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 177:58-65. [PMID: 30974244 DOI: 10.1016/j.ecoenv.2019.03.109] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 03/26/2019] [Accepted: 03/27/2019] [Indexed: 06/09/2023]
Abstract
Previous studies have demonstrated that the anti-androgenic effects of cypermethrin (CYP) are associated with testosterone (T) - related signaling pathway. This study was to investigate the effects of CYP on mouse Sertoli cells (TM4) and clarify whether the mechanisms were mediated by non-classical T signaling pathway activating mitogen-activated protein kinase (MAPK) cascade. The Cell Counting Kit 8 (CCK8) and Real-Time Cell Analysis iCELLigence (RTCA-iCELLigence) system were performed to detect the effects of 10 μM, 20 μM, 40 μM and 80 μM CYP on the viability and proliferation of TM4. The mammalian two hybrid assay, quantitative Real-Time PCR (qRT-PCR) and western blot were conducted to analyze the key genes and proteins involved in T-mediated MAPK signaling pathway. CYP was found to inhibit the viability and proliferation of TM4. Additionally, CYP disturbed the functions of Sertoli cells by inhibiting inhibin B (INH B) expression and facilitating androgen binding protein (ABP) and transferrin (TF) expression. Moreover, CYP suppressed the interaction of AR and Src kinase and inhibited androgen-mediated phosphorylation of Src, epidermal growth factor receptor (EGFR), extracellular-regulated kinase1/2 (ERK1/2) and transcription factor cAMP response element binding protein (CREB). Furthermore, the androgen-induced mRNA and protein expression of CREB-regulated gene early growth response factor (Egr1) decreased after treated with CYP. It is indicated that CYP inhibits the viability and proliferation of Sertoli cells and non-classical T signaling pathway activation of MAPK cascade is involved in anti-androgenic effect of CYP. This study provides a novel insight into the CYP-induced reproductive toxicity.
Collapse
Affiliation(s)
- Qi Wang
- School of Public Health, Xuzhou Medical University, Xuzhou, 209 Tong-Shan Road, Xuzhou, Jiangsu, 221002, China
| | - Heng-Xue Wang
- School of Public Health, Xuzhou Medical University, Xuzhou, 209 Tong-Shan Road, Xuzhou, Jiangsu, 221002, China
| | - Jun-Yu Shen
- School of Public Health, Xuzhou Medical University, Xuzhou, 209 Tong-Shan Road, Xuzhou, Jiangsu, 221002, China
| | - Rui Zhang
- School of Public Health, Xuzhou Medical University, Xuzhou, 209 Tong-Shan Road, Xuzhou, Jiangsu, 221002, China
| | - Jia-Wei Hong
- School of Public Health, Xuzhou Medical University, Xuzhou, 209 Tong-Shan Road, Xuzhou, Jiangsu, 221002, China
| | - Zheng Li
- School of Public Health, Xuzhou Medical University, Xuzhou, 209 Tong-Shan Road, Xuzhou, Jiangsu, 221002, China
| | - Gang Chen
- School of Public Health, Xuzhou Medical University, Xuzhou, 209 Tong-Shan Road, Xuzhou, Jiangsu, 221002, China
| | - Meng-Xue Li
- School of Public Health, Xuzhou Medical University, Xuzhou, 209 Tong-Shan Road, Xuzhou, Jiangsu, 221002, China
| | - Zhen Ding
- School of Public Health, Xuzhou Medical University, Xuzhou, 209 Tong-Shan Road, Xuzhou, Jiangsu, 221002, China
| | - Jing Li
- School of Public Health, Xuzhou Medical University, Xuzhou, 209 Tong-Shan Road, Xuzhou, Jiangsu, 221002, China
| | - Jin-Peng Zhang
- School of Public Health, Xuzhou Medical University, Xuzhou, 209 Tong-Shan Road, Xuzhou, Jiangsu, 221002, China
| | - Mei-Rong Zhang
- School of Public Health, Xuzhou Medical University, Xuzhou, 209 Tong-Shan Road, Xuzhou, Jiangsu, 221002, China
| | - Li-Chun Xu
- School of Public Health, Xuzhou Medical University, Xuzhou, 209 Tong-Shan Road, Xuzhou, Jiangsu, 221002, China.
| |
Collapse
|
37
|
Edelsztein NY, Rey RA. Importance of the Androgen Receptor Signaling in Gene Transactivation and Transrepression for Pubertal Maturation of the Testis. Cells 2019; 8:E861. [PMID: 31404977 PMCID: PMC6721648 DOI: 10.3390/cells8080861] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/30/2019] [Accepted: 08/01/2019] [Indexed: 12/27/2022] Open
Abstract
Androgens are key for pubertal development of the mammalian testis, a phenomenon that is tightly linked to Sertoli cell maturation. In this review, we discuss how androgen signaling affects Sertoli cell function and morphology by concomitantly inhibiting some processes and promoting others that contribute jointly to the completion of spermatogenesis. We focus on the molecular mechanisms that underlie anti-Müllerian hormone (AMH) inhibition by androgens at puberty, as well as on the role androgens have on Sertoli cell tight junction formation and maintenance and, consequently, on its effect on proper germ cell differentiation and meiotic onset during spermatogenesis.
Collapse
Affiliation(s)
- Nadia Y Edelsztein
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE) - CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires C1425EFD, Argentina.
| | - Rodolfo A Rey
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE) - CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires C1425EFD, Argentina.
- Departamento de Biología Celular, Histología, Embriología y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina.
| |
Collapse
|
38
|
|
39
|
Lim W, Ham J, Park S, Bae H, You S, Song G. Gossypol Induces Disruption of Spermatogenesis and Steroidogenesis in Male Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:2075-2085. [PMID: 30678458 DOI: 10.1021/acs.jafc.8b06946] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Gossypol, commonly found in cotton seeds, is hazardous to male reproductive physiology. Although several studies have indicated the toxicity of gossypol in human and animal reproduction, the mechanism of gossypol action in testes has not yet been elucidated. In the present study, we investigated the effects of gossypol in normal mouse testis cells, TM3 and TM4 cells, and in gossypol-treated C57BL/6 mice. We confirmed the antiproliferative effects of gossypol using cell viability assays, with PCNA as a proliferation marker, and cell cycle analysis. We also verified mitochondrial dysfunction and Ca2+ dysregulation in the cytosol of TM3 and TM4 cells, using JC-1 and Fluo-4 dyes. To confirm the cellular signaling mechanisms in testis cell lines, we performed Western blot analysis to assess the changes in MAPK and PI3K/Akt signal transduction, using their pharmacological inhibitors. Moreover, we screened the mRNA expression of genes involved in spermatogenesis and steroidogenesis in TM3 and TM4 cells. We also confirmed the mRNA expression and localization of genes regulating testis function in gossypol-treated and untreated mice testes. Collectively, we suggest that gossypol induces negative effects on testis function by reducing cell viability, mitochondrial membrane potential, and testis development-related genes in vitro and in vivo as well as by modulating the MAPK and PI3K signaling pathways.
Collapse
Affiliation(s)
- Whasun Lim
- Department of Food and Nutrition , Kookmin University , Seoul , 02707 , Republic of Korea
| | - Jiyeon Ham
- Department of Biotechnology , Korea University , Seoul , 02841 , Republic of Korea
| | - Sunwoo Park
- Department of Biotechnology , Korea University , Seoul , 02841 , Republic of Korea
| | - Hyocheol Bae
- Department of Biotechnology , Korea University , Seoul , 02841 , Republic of Korea
| | - Seungkwon You
- Department of Biotechnology , Korea University , Seoul , 02841 , Republic of Korea
| | - Gwonhwa Song
- Department of Biotechnology , Korea University , Seoul , 02841 , Republic of Korea
| |
Collapse
|
40
|
Bhattacharya I, Basu S, Pradhan BS, Sarkar H, Nagarajan P, Majumdar SS. Testosterone augments FSH signaling by upregulating the expression and activity of FSH-Receptor in Pubertal Primate Sertoli cells. Mol Cell Endocrinol 2019; 482:70-80. [PMID: 30579957 DOI: 10.1016/j.mce.2018.12.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 12/16/2018] [Accepted: 12/18/2018] [Indexed: 11/30/2022]
Abstract
The synergistic actions of Testosterone (T) and FSH via testicular Sertoli cells (Sc) regulate male fertility. We have previously reported that the actions of these hormones (T and FSH) in infant monkey testes are restricted only to the expansion of Sc and spermatogonial cells. The robust differentiation of male Germ cells (Gc) occurs after pubertal maturation of testis. The present study was aimed to investigate the molecular basis of the synergy between T and FSH action in pubertal primate (Macaca mulatta) Sc. Using primary Sc culture, we here have demonstrated that T (but not FSH) downregulated AMH and Inhibin-β-B (INHBB) mRNAs in pubertal Sc. We also found that, prolonged stimulation of T in pubertal Sc significantly elevated the expression of genes involved in FSH signaling pathway like FSH-Receptor (FSHR), GNAS and RIC8B, and this was associated with a rise in cAMP production. T also augmented FSH induced expression of genes like SCF, GDNF, ABP and Transferrin (TF) in pubertal Sc. We therefore conclude that T acts in synergy with FSH signaling in pubertal Sc. Such a coordinated network of hormonal signaling in Sc may facilitate the timely onset of the first spermatogenic wave in pubertal primates and is responsible for quantitatively and qualitatively normal spermatogenesis.
Collapse
Affiliation(s)
- Indrashis Bhattacharya
- Cellular Endocrinology Laboratory, National Institute of Immunology, New Delhi, 110067, India; Primate Research Center, National Institute of Immunology, New Delhi, India; Department of Zoology and Biotechnology, HNB Garhwal University, Srinagar Campus, Uttarakhand, India
| | - Sayon Basu
- Cellular Endocrinology Laboratory, National Institute of Immunology, New Delhi, 110067, India
| | - Bhola Shankar Pradhan
- Cellular Endocrinology Laboratory, National Institute of Immunology, New Delhi, 110067, India
| | - Hironmoy Sarkar
- Cellular Endocrinology Laboratory, National Institute of Immunology, New Delhi, 110067, India; Department of Microbiology, Raiganj University, Raiganj, West Bengal, India
| | - Perumal Nagarajan
- Primate Research Center, National Institute of Immunology, New Delhi, India
| | - Subeer S Majumdar
- Cellular Endocrinology Laboratory, National Institute of Immunology, New Delhi, 110067, India; Primate Research Center, National Institute of Immunology, New Delhi, India; National Institute of Animal Biotechnology, Hyderabad, Telangana, India.
| |
Collapse
|
41
|
Rodríguez-Lozano DC, Piña-Medina AG, Hansberg-Pastor V, Bello-Alvarez C, Camacho-Arroyo I. Testosterone Promotes Glioblastoma Cell Proliferation, Migration, and Invasion Through Androgen Receptor Activation. Front Endocrinol (Lausanne) 2019; 10:16. [PMID: 30778332 PMCID: PMC6369181 DOI: 10.3389/fendo.2019.00016] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 01/10/2019] [Indexed: 01/04/2023] Open
Abstract
Glioblastomas (GBM) are the most frequent and aggressive human brain tumors due to their high capacity to migrate and invade normal brain tissue. Epidemiological data report that GBM occur in a greater proportion in men than in women (3:2), suggesting the participation of sex hormones in the development of these tumors. It has been reported an increase in testosterone (T) levels in patients with GBM. In addition, androgen receptor (AR) is overexpressed in human GBM, and genetic silencing of AR, and its pharmacological inhibition, induce GBM cell death in vivo and in vitro. However, the role of T in proliferation, migration and invasion in human GBM cell lines has not been evaluated. We observed that T increased the number of U87, U251, and D54 cells derived from human GBM due to an increase in cell proliferation. This induction was blocked with flutamide, an antagonist of AR. T also induced migration and invasion of GBM cells that flutamide partially blocked. These data suggest that T through AR contributes to the progression of GBM by promoting proliferation, migration, and invasion.
Collapse
Affiliation(s)
- Dulce Carolina Rodríguez-Lozano
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Ana Gabriela Piña-Medina
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | - Claudia Bello-Alvarez
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Ignacio Camacho-Arroyo
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| |
Collapse
|
42
|
Meroni SB, Galardo MN, Rindone G, Gorga A, Riera MF, Cigorraga SB. Molecular Mechanisms and Signaling Pathways Involved in Sertoli Cell Proliferation. Front Endocrinol (Lausanne) 2019; 10:224. [PMID: 31040821 PMCID: PMC6476933 DOI: 10.3389/fendo.2019.00224] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 03/21/2019] [Indexed: 12/16/2022] Open
Abstract
Sertoli cells are somatic cells present in seminiferous tubules which have essential roles in regulating spermatogenesis. Considering that each Sertoli cell is able to support a limited number of germ cells, the final number of Sertoli cells reached during the proliferative period determines sperm production capacity. Only immature Sertoli cells, which have not established the blood-testis barrier, proliferate. A number of hormonal cues regulate Sertoli cell proliferation. Among them, FSH, the insulin family of growth factors, activin, and cytokines action must be highlighted. It has been demonstrated that cAMP/PKA, ERK1/2, PI3K/Akt, and mTORC1/p70SK6 pathways are the main signal transduction pathways involved in Sertoli cell proliferation. Additionally, c-Myc and hypoxia inducible factor are transcription factors which participate in the induction by FSH of various genes of relevance in cell cycle progression. Cessation of proliferation is a pre-requisite to Sertoli cell maturation accompanied by the establishment of the blood-testis barrier. With respect to this barrier, the participation of androgens, estrogens, thyroid hormones, retinoic acid and opioids has been reported. Additionally, two central enzymes that are involved in sensing cell energy status have been associated with the suppression of Sertoli cell proliferation, namely AMPK and Sirtuin 1 (SIRT1). Among the molecular mechanisms involved in the cessation of proliferation and in the maturation of Sertoli cells, it is worth mentioning the up-regulation of the cell cycle inhibitors p21Cip1, p27Kip, and p19INK4, and of the gap junction protein connexin 43. A decrease in Sertoli cell proliferation due to administration of certain therapeutic drugs and exposure to xenobiotic agents before puberty has been experimentally demonstrated. This review focuses on the hormones, locally produced factors, signal transduction pathways, and molecular mechanisms controlling Sertoli cell proliferation and maturation. The comprehension of how the final number of Sertoli cells in adulthood is established constitutes a pre-requisite to understand the underlying causes responsible for the progressive decrease in sperm production that has been observed during the last 50 years in humans.
Collapse
|
43
|
Kumar A, Dumasia K, Deshpande S, Raut S, Balasinor NH. Delineating the regulation of estrogen and androgen receptor expression by sex steroids during rat spermatogenesis. J Steroid Biochem Mol Biol 2018; 182:127-136. [PMID: 29709634 DOI: 10.1016/j.jsbmb.2018.04.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 04/18/2018] [Accepted: 04/26/2018] [Indexed: 12/24/2022]
Abstract
Estrogen receptors (ERα and β) and androgen receptor (AR) regulate various critical processes during spermatogenesis. Since spermatogenesis is very sensitive to hormonal stimuli and perturbations, it is important to understand the regulation of expression of these receptors by sex steroid hormones. Although many studies have reported deregulation of steroid receptors on endocrine disruption, there is no consensus on the regulation of their expression by steroid hormones during spermatogenesis, and a lack of clear understanding of the mechanism of regulation. Here, we evaluated the receptor expressions in a well-established exogenous estradiol administration model. We then investigated the mechanisms by which the individual receptors regulate their expression by binding to the respective hormone response elements upstream of these receptor genes. By further employing in vitro and in vivo models of ER and AR stimulation or antagonism, we delineated their regulation in a receptor subtype-specific manner. Our results indicate that ERα positively regulates expression of both the ERs; whereas, ERβ and AR negatively regulate expression of both ERβ and AR by direct binding to upstream regulatory regions. The perturbations in the levels of steroid receptors could be an important factor contributing to spermatogenic defects and male sub-fertility after estradiol and ER agonist treatment. Our study delineates the direct contribution of the individual steroid receptors in the regulation of their own expression.
Collapse
Affiliation(s)
- Anita Kumar
- Neuroendocrinology Division, National Institute for Research in Reproductive Health, Parel, Mumbai, 400012, India
| | - Kushaan Dumasia
- Neuroendocrinology Division, National Institute for Research in Reproductive Health, Parel, Mumbai, 400012, India
| | - Sharvari Deshpande
- Neuroendocrinology Division, National Institute for Research in Reproductive Health, Parel, Mumbai, 400012, India
| | - Sanketa Raut
- Neuroendocrinology Division, National Institute for Research in Reproductive Health, Parel, Mumbai, 400012, India
| | - N H Balasinor
- Neuroendocrinology Division, National Institute for Research in Reproductive Health, Parel, Mumbai, 400012, India.
| |
Collapse
|
44
|
Wang L, Song T, Wang X, Li J. Discovery and Identification of Pyrazolopyramidine Analogs as Novel Potent Androgen Receptor Antagonists. Front Pharmacol 2018; 9:864. [PMID: 30210333 PMCID: PMC6121070 DOI: 10.3389/fphar.2018.00864] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 07/17/2018] [Indexed: 01/21/2023] Open
Abstract
Androgen receptor (AR), an important target in the current androgen derivation therapy, plays a critical role in the development and progress of prostate cancer (PCa). Nonsteroidal antiandrogens, such as enzalutamide and bicalutamide, are commonly used in clinic to treat PCa. Though they are very effective at the beginning, drug resistance problem appears after about 18 months. One of the reasons is that these antiandrogens share similar structure skeleton. Therefore, it is urgent to discover novel antiandrogens with different skeletons for resistance problem. Herein, we combined structure- and ligand-based methodologies for virtual screening chemical databases to identify potent AR antagonists. Then the cytotoxic activities of the screened hit samples were evaluated by using LNCaP prostate cancer cells. Virtual screening and biological evaluation assay results suggest that several chemicals with novel pyrazolopyrimidine skeleton can inhibit the proliferation of prostate cancer cells with similar, or even higher, bioactivities to bicalutamide. AR reporter gene assay experiments proved that Compound III showed potential antagonistic effects. In addition, molecular dynamics simulations results proved that Compound III can properly bind to AR and prevent helix 12 (H12) from closing to distort the formation of activation function 2 (AF2) site, resulting in the invalid transcription. Hence, pyrazolopyrimidine was discovered as a novel, potent and promising antiandrogen skeleton deserved to be further studied.
Collapse
Affiliation(s)
- Lingyan Wang
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Tianqing Song
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Xin Wang
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Jiazhong Li
- School of Pharmacy, Lanzhou University, Lanzhou, China
| |
Collapse
|
45
|
Rajamanickam GD, Kastelic JP, Thundathil JC. The ubiquitous isoform of Na/K-ATPase (ATP1A1) regulates junctional proteins, connexin 43 and claudin 11 via Src-EGFR-ERK1/2-CREB pathway in rat Sertoli cells. Biol Reprod 2018; 96:456-468. [PMID: 28203706 DOI: 10.1095/biolreprod.116.141267] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 11/10/2016] [Accepted: 12/13/2016] [Indexed: 01/03/2023] Open
Abstract
Interaction of Na/K-ATPase with its ligand ouabain has been implicated in the regulation of various biological processes. The objective was to investigate roles of Na/K-ATPase isoforms in formation and function of junctional complexes in Sertoli cells. Primary cultures of Sertoli cells were obtained by enzymatic digestion of 20-day-old rat testes and grown on Matrigel-coated dishes for 7 days. Sertoli cells predominantly expressed the ubiquitous isoform of Na/K-ATPase (ATP1A1), confirmed by immunoblotting, PCR, immunofluorescence, and mass spectrometry. Treatment of Sertoli cells with 50 nM ouabain increased transepithelial electrical resistance (TER) and expression of claudin 11 (tight junctions) and connexin 43 (gap junctions), whereas 1 mM ouabain had opposite effects. Involvement of Src-EGFR-ERK1/2-CREB pathway in ouabain-mediated expression of claudin 11 and connexin 43 was evaluated. Incubation of Sertoli cells with 50 nM ouabain increased content of p-Src, p-EGFR, p-ERK1/2, and p-CREB; in contrast, 1 mM ouabain decreased phosphorylation of these signaling molecules. Preincubation of Sertoli cells with inhibitors of Src and MAPK pathways inhibited ouabain-induced effects on these signaling molecules, TER, and expression of claudin 11 and connexin 43. In conclusion, we inferred that ATP1A1 regulated Sertoli cell tight junctions and gap junctions through the Src-EGFR-ERK1/2-CREB pathway. Ouabain is an endogenous steroid; therefore, its interaction with ATP1A1 may be a critical signaling mechanism for the regulation of Sertoli cell function and male fertility.
Collapse
Affiliation(s)
- Gayathri D Rajamanickam
- Department of Production Animal Health, Faculty of Veterinary Medicine, Heritage Medical Research Building RM 400, 3330 Hospital Drive NW, University of Calgary, Calgary, AB, Canada
| | - John P Kastelic
- Department of Production Animal Health, Faculty of Veterinary Medicine, Heritage Medical Research Building RM 400, 3330 Hospital Drive NW, University of Calgary, Calgary, AB, Canada
| | - Jacob C Thundathil
- Department of Production Animal Health, Faculty of Veterinary Medicine, Heritage Medical Research Building RM 400, 3330 Hospital Drive NW, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
46
|
Hejmej A, Bilinska B. The effects of flutamide on cell-cell junctions in the testis, epididymis, and prostate. Reprod Toxicol 2018; 81:1-16. [PMID: 29958919 DOI: 10.1016/j.reprotox.2018.06.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 06/15/2018] [Accepted: 06/20/2018] [Indexed: 12/12/2022]
Abstract
In this review, we summarize recent findings on the effect of the anti-androgen flutamide on cell-cell junctions in the male reproductive system. We outline developmental aspects of flutamide action on the testis, epididymis, and prostate, and describe changes in junction protein expression and organization of junctional complexes in the adult boar following prenatal and postnatal exposure. We also discuss findings on the mechanisms by which flutamide induces alterations in cell-cell junctions in reproductive tissues of adult males, with special emphasis on cytoplasmic effects. Based on the results from in vivo and in vitro studies in the rat, we propose that flutamide affects the expression of junction proteins and junction complex structure not only by inhibiting androgen receptor activity, but equally important by modulating protein kinase-dependent signaling in testicular cells. Additionally, results from studies on prostate cancer cell lines point to a role for the cellular molecular outfit in response to flutamide.
Collapse
Affiliation(s)
- Anna Hejmej
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Barbara Bilinska
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland.
| |
Collapse
|
47
|
Schreihofer DA, Duong P, Cunningham RL. N-terminal truncations in sex steroid receptors and rapid steroid actions. Steroids 2018; 133:15-20. [PMID: 29104096 PMCID: PMC5864524 DOI: 10.1016/j.steroids.2017.10.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 10/27/2017] [Accepted: 10/30/2017] [Indexed: 01/14/2023]
Abstract
Sex steroid receptors act as ligand activated nuclear transcription factors throughout the body, including the brain. However, post-translational modification of these receptors can direct them to extranuclear sites, including the plasma membrane, where they are able to initiate rapid signaling. Because of the conserved domain structure of these receptors, alternative exon splicing can result in proteins with altered nuclear and extranuclear actions. Although much attention has focused on internal and C-terminal splice variants, both estrogen and androgen receptors undergo N-terminal truncations, as well. These truncated proteins not only influence the transcriptional activity of the full-length receptors, but also associate with caveolin and initiate signaling at the plasma membrane. Such actions may have important physiological consequences in neuronal, endothelial, and cancer signaling and cell survival.
Collapse
Affiliation(s)
- Derek A Schreihofer
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie Boulevard, Fort Worth, TX 76244, USA
| | - Phong Duong
- Department of Physiology and Anatomy, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie Boulevard, Fort Worth, TX 76244, USA
| | - Rebecca L Cunningham
- Department of Physiology and Anatomy, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie Boulevard, Fort Worth, TX 76244, USA.
| |
Collapse
|
48
|
Salerno M, Cascio O, Bertozzi G, Sessa F, Messina A, Monda V, Cipolloni L, Biondi A, Daniele A, Pomara C. Anabolic androgenic steroids and carcinogenicity focusing on Leydig cell: a literature review. Oncotarget 2018; 9:19415-19426. [PMID: 29721213 PMCID: PMC5922407 DOI: 10.18632/oncotarget.24767] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 02/27/2018] [Indexed: 01/25/2023] Open
Abstract
Anabolic androgenic steroids (AAS) are some of the most common drugs used among athletes, frequently in combination with resistance training, to improve physical performance or for aesthetic purpose. A great number of scientific reports showed the detrimental effects of anabolic androgenic steroids on different organs and tissues. In this literature review, we analyzed the AAS-mediated carcinogenicity, focusing on Leydig cell tumor. AAS-induced carcinogenicity can affect DNA transcription through two pathways. It can act directly via the androgen receptor, by means of dihydrotestosterone (DHT) produced by the action of 5-a-reductase. It can also work through the estrogen receptor, by means of estradiol produced by CYP19 aromatase. In addition, nandrolone and stanazolol can activate the PI3K/AKT and PLC/PKC pathways via IGF-1. This would result in cell proliferation in Leydig cell cancer, or magnify cyclin D1 concentration inducing breast cell proliferation. AAS abuse is becoming a serious public health concern in view of the severe health consequences secondary to AAS abuse. The negative role of AAS in supraphysiological dosage impairs the expression of enzymes involved in testosterone biosynthesis. Abnormal synthesis of testosterone plays has a negative effect on the hormonal changes/regulation, and might be involved in certain carcinogenic mechanisms. At the light of this review, it could become very interesting to perform an information campaign more strengthened in gyms and schools in order to prevent male fertility impairment and other tissues damage.
Collapse
Affiliation(s)
- Monica Salerno
- University of Foggia, Department of Clinical and Experimental Medicine, Foggia, Italy
| | - Orazio Cascio
- University of Catania, Department of Medical, Surgical and Advanced Technologies, "G.F. Ingrassia", Catania, Italy
| | - Giuseppe Bertozzi
- University of Foggia, Department of Clinical and Experimental Medicine, Foggia, Italy
| | - Francesco Sessa
- University of Foggia, Department of Clinical and Experimental Medicine, Foggia, Italy
| | - Antonietta Messina
- University of Campania "L. Vanvitelli", Department of Experimental Medicine, Naples, Italy
| | - Vincenzo Monda
- University of Campania "L. Vanvitelli", Department of Experimental Medicine, Naples, Italy
| | - Luigi Cipolloni
- Università degli Studi di Roma "La Sapienza", Department of Public Health, Roma, Italy
| | - Antonio Biondi
- University of Catania, Department of Surgery, Catania, Italy
| | - Aurora Daniele
- University of Campania "L. Vanvitelli", CEINGE Biotecnologie Avanzate S.C. a r.l., Naples, Italy
| | - Cristoforo Pomara
- University of Catania, Department of Medical, Surgical and Advanced Technologies, "G.F. Ingrassia", Catania, Italy
| |
Collapse
|
49
|
Gegenschatz-Schmid K, Verkauskas G, Demougin P, Bilius V, Dasevicius D, Stadler MB, Hadziselimovic F. Curative GnRHa treatment has an unexpected repressive effect on Sertoli cell specific genes. Basic Clin Androl 2018; 28:2. [PMID: 29456864 PMCID: PMC5806254 DOI: 10.1186/s12610-018-0067-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 01/23/2018] [Indexed: 12/27/2022] Open
Abstract
Background Follicle stimulating hormone and testosterone stimulate Sertoli cells to support germ cell function and differentiation. During mini-puberty, when gonadotropin (GnRH) stimulates increases in plasma luteinizing hormone (LH) and testosterone levels, gonocytes are transformed into Ad spermatogonia. In cryptorchidism, impaired gonadotropin secretion during mini-puberty results in insufficient LH and testosterone secretion, impaired gonocyte transition to Ad spermatogonia, and perturbed Sertoli cell proliferation. Treatment with a gonadotropin-releasing hormone agonist (GnRHa/Buserelin) induced gonocytes to differentiate into Ad spermatogonia and rescued fertility. The present study evaluated the impact of low LH secretion on Sertoli cell function by comparing differential gene expression data between testes with low LH that lacked Ad spermatogonia (Ad-) and testes that completed mini-puberty (Ad+). Furthermore, we analyzed changes in the transcription of selected Sertoli cell specific genes in response to GnRHa treatment. Results Ad- testes showed reduced expression of nine out of 40 selected Sertoli cell specific genes compared to Ad+ testes. GnRHa treatment repressed most of the Sertoli cell specific genes, including the inhibins, but it increased the expression of genes that regulate apoptosis (FASLG) and proliferation (GDNF). Conclusions Impaired-minipuberty with decreased LH and testosterone levels affected Ad and Sertoli cell development through positive and negative regulation of morphoregulatory and apoptotic genes. GnRHa treatment had a repressive effect on most Sertoli cell specific genes, which suggested that Sertoli cells underwent a cellular rearrangement. We propose that gonadotropin-dependent increases in FASLG and GDNF expression drove Sertoli cell proliferation and germ cell self-renewal and supported the transition of gonocytes to Ad spermatogonia, independent of inhibins.
Collapse
Affiliation(s)
| | - Gilvydas Verkauskas
- 2Children's Surgery Centre, Faculty of Medicine, Vilnius University, 01513 Vilnius, Lithuania
| | - Philippe Demougin
- 3Biozentrum, Life Sciences Training Facility, University of Basel, 4001 Basel, Switzerland
| | - Vytautas Bilius
- 2Children's Surgery Centre, Faculty of Medicine, Vilnius University, 01513 Vilnius, Lithuania
| | - Darius Dasevicius
- 4Institute for Pathology, National Centre of Pathology, Affiliate of Vilnius University Hospital Santariskiu Klinikos, 08406 Vilnius, Lithuania
| | - Michael B Stadler
- 5Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland.,6Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Faruk Hadziselimovic
- Cryptorchidism Research Institute, Kindermedizinisches Zentrum Liestal, 4410 Liestal, Switzerland
| |
Collapse
|
50
|
Thomas P, Converse A, Berg HA. ZIP9, a novel membrane androgen receptor and zinc transporter protein. Gen Comp Endocrinol 2018; 257:130-136. [PMID: 28479083 DOI: 10.1016/j.ygcen.2017.04.016] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 04/11/2017] [Accepted: 04/29/2017] [Indexed: 11/17/2022]
Abstract
Rapid, androgen actions initiated at the cell surface have been reported in a variety of vertebrate cells, including several macrophage and prostate cancer cell lines that lack the nuclear androgen receptor. However, until recently the identity of the novel membrane androgen receptor (mAR) mediating these nonclassical androgen actions remained unknown. In 2014, a novel mAR unrelated to nuclear androgen receptors was identified in Atlantic croaker ovaries as the zinc transporter protein, ZIP9. ZIP9 is one of the 14 members of the ZIP (ZRT-and Irt-like Protein, SLC39A) family that regulates zinc homeostasis by transporting zinc across cell and organelle membranes into the cytoplasm. Zinc is a micronutrient critical for the maintenance of physiological and cellular processes, such as development, growth, protein assembly and activity, signaling, and apoptosis. Both croaker ZIP9 and human ZIP9 proteins have the binding characteristics of high affinity, specific mARs, and are coupled to G proteins. Testosterone induces apoptosis through ZIP9 in croaker granulosa cells and in human breast and prostate cancer cells by a unique mechanism involving increases in both second messengers and intracellular free zinc concentrations. ZIP9 also mediates testosterone regulation of tight junction formation in Sertoli cells and nonclassical testosterone signaling in spermatogenic cells. ZIP9 acts through several signal transduction pathways, a stimulatory G protein (Gs) in granulosa cells, an inhibitory one (Gi) in cancer cells, and a Gq11 one (Gnα11) in spermatogenic cells. ZIP9 has a very broad tissue distribution and is predicted to mediate numerous and diverse nonclassical androgen actions in vertebrates.
Collapse
Affiliation(s)
- Peter Thomas
- Marine Science Institute, The University of Texas at Austin, 750 Channel View Drive, Port Aransas, TX 78373, United States.
| | - Aubrey Converse
- Marine Science Institute, The University of Texas at Austin, 750 Channel View Drive, Port Aransas, TX 78373, United States
| | - Håkan A Berg
- Department of Science and Technology, Örebro University, Örebro, Sweden
| |
Collapse
|