1
|
Augière C, Campolina-Silva G, Vijayakumaran A, Medagedara O, Lavoie-Ouellet C, Joly Beauparlant C, Droit A, Barrachina F, Ottino K, Battistone MA, Narayan K, Hess R, Mennella V, Belleannée C. ARL13B controls male reproductive tract physiology through primary and Motile Cilia. Commun Biol 2024; 7:1318. [PMID: 39397107 PMCID: PMC11471856 DOI: 10.1038/s42003-024-07030-7] [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: 04/09/2024] [Accepted: 10/07/2024] [Indexed: 10/15/2024] Open
Abstract
ARL13B is a small regulatory GTPase that controls ciliary membrane composition in both motile cilia and non-motile primary cilia. In this study, we investigated the role of ARL13B in the efferent ductules, tubules of the male reproductive tract essential to male fertility in which primary and motile cilia co-exist. We used a genetically engineered mouse model to delete Arl13b in efferent ductule epithelial cells, resulting in compromised primary and motile cilia architecture and functions. This deletion led to disturbances in reabsorptive/secretory processes and triggered an inflammatory response. The observed male reproductive phenotype showed significant variability linked to partial infertility, highlighting the importance of ARL13B in maintaining a proper physiological balance in these small ducts. These results emphasize the dual role of both motile and primary cilia functions in regulating efferent duct homeostasis, offering deeper insights into how cilia related diseases affect the male reproductive system.
Collapse
Affiliation(s)
- Céline Augière
- CHU de Québec Research Center (CHUL)- Université Laval, Quebec City, QC, Canada.
- Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle, Department of Obstetrics, Gynecology, and Reproduction, Faculty of Medicine, Université Laval, Quebec City, QC, Canada.
| | - Gabriel Campolina-Silva
- CHU de Québec Research Center (CHUL)- Université Laval, Quebec City, QC, Canada
- Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle, Department of Obstetrics, Gynecology, and Reproduction, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Aaran Vijayakumaran
- Medical Research Council Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge, UK
| | - Odara Medagedara
- Medical Research Council Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge, UK
| | - Camille Lavoie-Ouellet
- CHU de Québec Research Center (CHUL)- Université Laval, Quebec City, QC, Canada
- Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle, Department of Obstetrics, Gynecology, and Reproduction, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | | | - Arnaud Droit
- CHU de Québec Research Center (CHUL)- Université Laval, Quebec City, QC, Canada
| | - Ferran Barrachina
- Program in Membrane Biology, Nephrology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, MA, USA
| | - Kiera Ottino
- Program in Membrane Biology, Nephrology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, MA, USA
| | - Maria Agustina Battistone
- Program in Membrane Biology, Nephrology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, MA, USA
| | - Kedar Narayan
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Rex Hess
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana, Illinois, IL, USA
| | - Vito Mennella
- Medical Research Council Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge, UK
- Department of Pathology, 10 Tennis Court Road, University of Cambridge, Cambridge, UK
| | - Clémence Belleannée
- CHU de Québec Research Center (CHUL)- Université Laval, Quebec City, QC, Canada.
- Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle, Department of Obstetrics, Gynecology, and Reproduction, Faculty of Medicine, Université Laval, Quebec City, QC, Canada.
| |
Collapse
|
2
|
Hess RA, Park CJ, Soto S, Reinacher L, Oh JE, Bunnell M, Ko CJ. Male animal sterilization: history, current practices, and potential methods for replacing castration. Front Vet Sci 2024; 11:1409386. [PMID: 39027909 PMCID: PMC11255590 DOI: 10.3389/fvets.2024.1409386] [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: 03/29/2024] [Accepted: 06/10/2024] [Indexed: 07/20/2024] Open
Abstract
Sterilization and castration have been synonyms for thousands of years. Making an animal sterile meant to render them incapable of producing offspring. Castration or the physical removal of the testes was discovered to be the most simple but reliable method for managing reproduction and sexual behavior in the male. Today, there continues to be global utilization of castration in domestic animals. More than six hundred million pigs are castrated every year, and surgical removal of testes in dogs and cats is a routine practice in veterinary medicine. However, modern biological research has extended the meaning of sterilization to include methods that spare testis removal and involve a variety of options, from chemical castration and immunocastration to various methods of vasectomy. This review begins with the history of sterilization, showing a direct link between its practice in man and animals. Then, it traces the evolution of concepts for inducing sterility, where research has overlapped with basic studies of reproductive hormones and the discovery of testicular toxicants, some of which serve as sterilizing agents in rodent pests. Finally, the most recent efforts to use the immune system and gene editing to block hormonal stimulation of testis function are discussed. As we respond to the crisis of animal overpopulation and strive for better animal welfare, these novel methods provide optimism for replacing surgical castration in some species.
Collapse
Affiliation(s)
- Rex A. Hess
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Epivara, Inc, Champaign, IL, United States
| | - Chan Jin Park
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Epivara, Inc, Champaign, IL, United States
| | | | | | - Ji-Eun Oh
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Mary Bunnell
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - CheMyong J. Ko
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Epivara, Inc, Champaign, IL, United States
| |
Collapse
|
3
|
Oh Y, Kasu M, Bottoms CJ, Douglas JC, Sekulovski N, Hayashi K, MacLean II JA. Rhox8 homeobox gene ablation leads to rete testis abnormality and male subfertility in mice†. Biol Reprod 2023; 109:520-532. [PMID: 37471646 PMCID: PMC10577278 DOI: 10.1093/biolre/ioad077] [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: 02/23/2023] [Revised: 07/10/2023] [Accepted: 07/15/2023] [Indexed: 07/22/2023] Open
Abstract
The reproductive homeobox X-linked (Rhox) genes encode transcription factors that are expressed selectively in reproductive tissues including the testis, epididymis, ovary, and placenta. While many Rhox genes are expressed in germ cells in the mouse testis, only Rhox8 is expressed exclusively in the Sertoli cells during embryonic and postnatal development, suggesting a possible role of Rhox8 in embryonic gonad development. Previously, Sertoli cell-specific knockdown of RHOX8 resulted in male subfertility due to germ cell defects. However, this knockdown model was limited in examining the functions of Rhox8 as RHOX8 knockdown occurred only postnatally, and there was still residual RHOX8 in the testis. In this study, we generated new Rhox8 knockout (KO) mice using the CRISPR/Cas9 system. Sex determination and fetal testis development were apparently normal in mutant mice. Fertility analysis showed a low fecundity in Rhox8 KO adult males, with disrupted spermatogenic cycles, increased germ cell apoptosis, and reduced sperm count and motility. Interestingly, Rhox8 KO testes showed an increase in testis size with dilated seminiferous tubules and rete testis, which might be affected by efferent duct (ED) Rhox8 ablation dysregulating the expression of metabolism and transport genes in the EDs. Taken together, the data presented in this study suggest that Rhox8 in the Sertoli cells is not essential for sex determination and embryonic testis differentiation but has an important role in complete spermatogenesis and optimal male fertility.
Collapse
Affiliation(s)
- Yeongseok Oh
- Center for Reproductive Biology, School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
- Department of Physiology, Southern Illinois School of Medicine, Carbondale, IL, USA
| | - Maho Kasu
- Center for Reproductive Biology, School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Constence J Bottoms
- Center for Reproductive Biology, School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Jenna C Douglas
- Center for Reproductive Biology, School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Nikola Sekulovski
- Department of Physiology, Southern Illinois School of Medicine, Carbondale, IL, USA
| | - Kanako Hayashi
- Center for Reproductive Biology, School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
- Department of Physiology, Southern Illinois School of Medicine, Carbondale, IL, USA
| | - James A MacLean II
- Center for Reproductive Biology, School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
- Department of Physiology, Southern Illinois School of Medicine, Carbondale, IL, USA
| |
Collapse
|
4
|
Omotehara T, Hess RA, Nakata H, Birch LA, Prins GS, Itoh M. Expression patterns of sex steroid receptors in developing mesonephros of the male mouse: three-dimensional analysis. Cell Tissue Res 2023; 393:577-593. [PMID: 37335379 DOI: 10.1007/s00441-023-03796-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 06/05/2023] [Indexed: 06/21/2023]
Abstract
The androgen pathway via androgen receptor (AR) has received the most attention for development of male reproductive tracts. The estrogen pathway through estrogen receptor (ESR1) is also a major contributor to rete testis and efferent duct formation, but the role of progesterone via progesterone receptor (PGR) has largely been overlooked. Expression patterns of these receptors in the mesonephric tubules (MTs) and Wolffian duct (WD), which differentiate into the efferent ductules and epididymis, respectively, remain unclear because of the difficulty in distinguishing each region of the tracts. This study investigated AR, ESR1, and PGR expressions in the murine mesonephros using three-dimensional (3-D) reconstruction. The receptors were localized in serial paraffin sections of the mouse testis and mesonephros by immunohistochemistry on embryonic days (E) 12.5, 15.5, and 18.5. Specific regions of the developing MTs and WD were determined by 3-D reconstruction using Amira software. AR was found first in the specific portion of the MTs near the MT-rete junction at E12.5, and the epithelial expression showed increasing strength from cranial to the caudal regions. Epithelial expression of ESR1 was found in the cranial WD and MTs near the WD first at E15.5. PGR was weakly positive only in the MTs and cranial WD starting on E15.5. This 3-D analysis suggests that gonadal androgen acts first on the MTs near the MT-rete junction but that estrogen is the first to influence MTs near the WD, while potential PGR activity is delayed and limited to the epithelium.
Collapse
Affiliation(s)
- Takuya Omotehara
- Department of Anatomy and Life Structure, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan.
- Department of Anatomy, Tokyo Medical University, Tokyo, Japan.
| | - Rex A Hess
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Chicago, IL, 61821, USA
| | - Hiroki Nakata
- Department of Clinical Engineering, Faculty of Health Sciences, Komatsu University, Komatsu, Japan
| | - Lynn A Birch
- Department of Urology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Gail S Prins
- Department of Urology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Masahiro Itoh
- Department of Anatomy, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
5
|
Zhou F, Guo C, Wang L, Zhang G, Wang J, Chen W, Cui K, Tan Y, Zhou Z. Mono-(2-ethylhexyl) Phthalate (MEHP)-Induced Telomere Structure and Function Disorder Mediates Cell Cycle Dysregulation and Apoptosis via c-Myc and Its Upstream Transcription Factors in a Mouse Spermatogonia-Derived (GC-1) Cell Line. TOXICS 2023; 11:toxics11050448. [PMID: 37235262 DOI: 10.3390/toxics11050448] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 04/30/2023] [Accepted: 05/06/2023] [Indexed: 05/28/2023]
Abstract
As a typical environmental endocrine disrupting chemical (EDC), di-(2-ethylhexyl) phthalate (DEHP) is thought to be related to reproductive disorders, especially in males. Growing evidence suggests that various EDCs may result in an impaired telomere structure and function, which is associated with male infertility. However, the adverse effect of DEHP on telomeres in male reproductive cells has rarely been studied, and the related mechanisms remain unclear. In this study, we tested the effects of mono-(2-ethylhexyl) phthalate (MEHP), the primary metabolite of DEHP, on telomere dysfunction in mouse spermatogonia-derived cells (GC-1) and the potential role of TERT and c-Myc in MEHP-induced spermatogenic cell damage. Results showed that MEHP induced cell viability inhibition, G0/G1 phase cell cycle arrest, and apoptosis in GC-1 cells in a dose-dependent manner. Shortened telomeres, reduced telomerase activity, and decreased expression of TERT, c-Myc, and upstream transcription factors of c-Myc were also observed in the MEHP-treated cells. In conclusion, it can be concluded that TERT-mediated telomere dysfunction may contribute to MEHP-induced G0/G1 phase cell cycle arrest and apoptosis in GC-1 cells through the impairment of c-Myc and its upstream transcription factors.
Collapse
Affiliation(s)
- Fangji Zhou
- Department of Environmental Health, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Chengwei Guo
- Department of Environmental Health, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Lingqiao Wang
- Department of Environmental Health, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Guowei Zhang
- Department of Environmental Health, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Jia Wang
- Department of Environmental Health, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Weiyan Chen
- Department of Environmental Health, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Ke Cui
- Department of Environmental Health, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yao Tan
- Department of Environmental Health, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Ziyuan Zhou
- Department of Environmental Health, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| |
Collapse
|
6
|
da Silva AAS, de Santi F, Hinton BT, Cerri PS, Sasso-Cerri E. Venlafaxine increases aromatization, reduces apical V-ATPase in clear cells and induces increased number of mast cells and smooth muscle cells death in rat cauda epididymis. Life Sci 2023; 315:121329. [PMID: 36584913 DOI: 10.1016/j.lfs.2022.121329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/12/2022] [Accepted: 12/23/2022] [Indexed: 12/29/2022]
Abstract
Depressive disorders (DD) have affected millions of people worldwide. Venlafaxine, antidepressant of the class of serotonin and norepinephrine reuptake inhibitors, has been prescribed for the treatment of DD. In rat testes, venlafaxine induces testosterone (T) aromatization and increases estrogen levels. Aromatase is a key enzyme for the formation of estrogen in the epididymis, an essential organ for male fertility. We investigated the impact of serotonergic/noradrenergic venlafaxine effect on the epididymal cauda region, focusing on aromatase, V-ATPase and EGF epithelial immunoexpression, smooth muscle (SM) integrity and mast cells number (MCN). Male rats were distributed into control (CG; n = 10) and venlafaxine (VFG, n = 10) groups. VFG received 30 mg/kg b.w. of venlafaxine for 35 days. The epididymal cauda was processed for light and transmission electron microscopy (TEM). The expression of connexin 43 (Cx43) and estrogen alpha (Esr1), adrenergic (Adra1a) and serotonergic (Htr1b) receptors were analyzed. Clear cells (CCs) area, SM thickness, viable spermatozoa (VS) and MCN were evaluated. Apoptosis was confirmed by TUNEL and TEM. The following immunoreactions were performed: T, aromatase, T/aromatase co-localization, V-ATPase, EGF, Cx43 and PCNA. The increased Adra1a and reduced Htr1b expressions confirmed the noradrenergic and serotonergic venlafaxine effects, respectively, corroborating the increased MCN, apoptosis and atrophy of SM. In VFG, the epithelial EGF increased, explaining Cx43 overexpression and basal cells mitotic activity. T aromatization and Esr1 downregulation indicate high estrogen levels, explaining CCs hypertrophy and changes in the V-ATPase localization, corroborating VS reduction. Thus, in addition to serotonergic/noradrenergic effects, T/estrogen imbalance, induced by venlafaxine, impairs epididymal structure and function.
Collapse
Affiliation(s)
- André Acácio Souza da Silva
- São Paulo State University (Unesp), School of Dentistry, Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry, Araraquara, Brazil
| | - Fabiane de Santi
- Federal University of São Paulo, Department of Morphology and Genetics, São Paulo, Brazil
| | - Barry T Hinton
- University of Virginia, School of Medicine, Department of Cell Biology, Charlottesville, USA
| | - Paulo Sérgio Cerri
- São Paulo State University (Unesp), School of Dentistry, Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry, Araraquara, Brazil
| | - Estela Sasso-Cerri
- São Paulo State University (Unesp), School of Dentistry, Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry, Araraquara, Brazil.
| |
Collapse
|
7
|
Chan CJ, Hirashima T. Tissue hydraulics in reproduction. Semin Cell Dev Biol 2022; 131:124-133. [PMID: 35606275 DOI: 10.1016/j.semcdb.2022.05.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 12/14/2022]
Abstract
The development of functional eggs and sperm are critical processes in mammalian development as they ensure successful reproduction and species propagation. While past studies have identified important genes that regulate these processes, the roles of luminal flow and fluid stress in reproductive biology remain less well understood. Here, we discuss recent evidence that support the diverse functions of luminal fluid in oogenesis, spermatogenesis and embryogenesis. We also review emerging techniques that allow for precise quantification and perturbation of tissue hydraulics in female and male reproductive systems, and propose new questions and approaches in this field. We hope this review will provide a useful resource to inspire future research in tissue hydraulics in reproductive biology and diseases.
Collapse
Affiliation(s)
- Chii Jou Chan
- Mechanobiology Institute, National University of Singapore, Singapore; Department of Biological Sciences, National University of Singapore, Singapore.
| | - Tsuyoshi Hirashima
- Mechanobiology Institute, National University of Singapore, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; The Hakubi Center/Graduate School of Biostudies, Kyoto University, Kyoto, Japan; Japan Science and Technology Agency, PRESTO, Kawaguchi, Japan.
| |
Collapse
|
8
|
The Mechanism and Experimental Validation of Forsythoside A in the Treatment of Male Infertility Were Analyzed Based on Network Pharmacology and Molecular Docking. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:7723358. [PMID: 36248414 PMCID: PMC9560825 DOI: 10.1155/2022/7723358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 08/17/2022] [Accepted: 08/24/2022] [Indexed: 11/15/2022]
Abstract
Chinese medicine extracts are currently the hotspot of new drug research and development. Herein, we report the mechanism of action of the traditional Chinese medicine extract Forsythiaside A in the treatment of male infertility and experimental verification. We first obtained 95 intersection genes between the target protein of Forsythiaside A and the target genes of male infertility and screened 13 key genes. In molecular docking, Forsythiaside A can each have a higher total docking score with 12 key genes and have a better combination. These 95 intersection genes are mainly related to biological processes such as response to peptide hormone, response to oxidative stress, and participation in the oxidative stress of the forkhead box O (FoxO) signaling pathway. Therefore, we use ornidazole to induce an experimental model of oligoasthenospermia in rats and use different concentrations of Forsythiaside A to intervene. We proved that the semen quality and superoxide dismutase (SOD) activities of model group rats were significantly lower than those of the blank group, and semen quality and SOD activities of the low-dose group and high-dose group were significantly higher than those of the model group. The malondialdehyde (MDA) level of model group rats was significantly higher than that of blank group, while the MDA levels of the low-dose group and high-dose group were significantly lower than that of the model group. Forsythoside A is a potential drug substance for male infertility and improves the semen quality, MDA levels, and SOD activities of rats with oligoasthenospermia.
Collapse
|
9
|
Sobhani K, Vaziry A, Farzinpour A, Rostamzadeh J. Relative expression of aromatase in the male goat reproductive organs during different seasons. Reprod Domest Anim 2022; 57:806-812. [DOI: 10.1111/rda.14118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/27/2022] [Indexed: 11/26/2022]
Affiliation(s)
- Keyvan Sobhani
- Department of Animal Science Faculty of Agriculture University of Kurdistan Sanandaj Kurdistan Iran
| | - Asaad Vaziry
- Department of Animal Science Faculty of Agriculture University of Kurdistan Sanandaj Kurdistan Iran
| | - Amjad Farzinpour
- Department of Animal Science Faculty of Agriculture University of Kurdistan Sanandaj Kurdistan Iran
| | - Jalal Rostamzadeh
- Department of Animal Science Faculty of Agriculture University of Kurdistan Sanandaj Kurdistan Iran
| |
Collapse
|
10
|
Chen R, He J, Li Y, An L, Hu J. Tricresyl phosphate inhibits fertilization in Japanese medaka (Oryzias latipes): Emphasizing metabolic toxicity. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 297:118809. [PMID: 35016985 DOI: 10.1016/j.envpol.2022.118809] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 12/19/2021] [Accepted: 01/05/2022] [Indexed: 06/14/2023]
Abstract
As tricresyl phosphate (TCrP) is commonly found in global water sources, its potential reproductive toxicity to fish is of increasing concern. Japanese medaka larvae were exposed to TCrP at 657.9, 1,511, and 4042 ng/L for 100 days. We identified significant fertilization inhibition (6.9%-12.8%) in all exposure groups. Intersex was significantly induced at 4042 ng/L, with an incidence of 22.0%. TCrP exposure also caused dilation of the efferent duct in the testes with maximum duct widths of 83.3, 93.2, and 149.7 μm in the 657.9, 1,511, and 4042 ng/L exposure groups, respectively. These widths were all significantly larger than that observed in the control group (37.7 μm) and likely contributed substantially to fertilization inhibition. The TCrP metabolites 4-OH-MDTP and 3-OH-MDTP, were detected at high concentrations in the liver and elicited 5.8-fold and 5.3-fold greater androgen receptor antagonistic activity than that elicited by TCrP (39.8 μM), which may explain the intersex observed in low exposure groups. 4-OH-MDTP and 3-OH-MDTP elicited anti-estrogenic activities by blocking the estrogen receptor, and the concentrations at which its responses were equal to the IC20 of tamoxifen were 16.1 μM and 18.9 μM, respectively, as detected using the yeast two-hybrid assay. Such anti-estrogenic activities were likely the main driver of dilation of the efferent duct. Observed adverse outcomes after exposure to TCrP all occurred under environmentally relevant concentrations, suggesting considerable ecological risk to wild fish.
Collapse
Affiliation(s)
- Ruichao Chen
- MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, China
| | - Jianwu He
- MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, China
| | - Yu Li
- State Environmental Protection Key Laboratory of Integrated Surface Water-Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Lihui An
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China
| | - Jianying Hu
- MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|
11
|
Pilutin A, Misiakiewicz-Has K, Rzeszotek S, Wiszniewska B. Morphological and morphometric changes and epithelial apoptosis are induced in the rat epididymis by long-term letrozole treatment. Eur J Histochem 2021; 65:3259. [PMID: 34474552 PMCID: PMC8431869 DOI: 10.4081/ejh.2021.3259] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 07/20/2021] [Indexed: 11/24/2022] Open
Abstract
The epididymis is an organ that plays a key role in sperm maturation. The aim of this study was to examine the association between the chronic treatment of mature male rats with letrozole and morphological evaluation and morphometric values of epididymis as well as changes in the number of apoptotic cells in epididymal epithelium. Adult rats were treated with letrozole for 6 months and the epididymis weight, morphology, morphometric values and the number of apoptotic cells in the epithelium were examined. Long-term aromatase inhibition resulted in presence of intraepithelial clear vacuoles, hyperplasia of clear cells and a hyperplastic alteration in the epithelium known as a cribriform change. Moreover, changes in diameters of the epididymal duct and the epididymal lumen and changes in the epididymal epithelium height were observed. The number of apoptotic epithelial cells was increased in letrozole-treated group. It can be indicated that chronic treatment with letrozole can affect morphology, morphometric values and apoptosis in the epididymis of adult male rats. Observed changes are similar to that observed in the aging processes and may also be important for patients treated with aromatase inhibitors.
Collapse
Affiliation(s)
- Anna Pilutin
- Department of Histology and Embryology, Pomeranian Medical University, Szczecin.
| | | | - Sylwia Rzeszotek
- Department of Histology and Embryology, Pomeranian Medical University, Szczecin.
| | - Barbara Wiszniewska
- Department of Histology and Embryology, Pomeranian Medical University, Szczecin.
| |
Collapse
|
12
|
Yang FW, Fang B, Pang GF, Zhang M, Ren FZ. Triazophos and its metabolite diethyl phosphate have different effects on endocrine hormones and gut health in rats. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART. B, PESTICIDES, FOOD CONTAMINANTS, AND AGRICULTURAL WASTES 2021; 56:566-576. [PMID: 34038317 DOI: 10.1080/03601234.2021.1922042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Organophosphorus pesticide (OP) residues present in food can be metabolized into diethylphosphate (DEP) in vivo. Epidemiological studies of OPs have usually focused on these metabolites, while animal studies mainly assessed the OPs. Here, we compared the health risks of a frequently detected OP, triazophos (TAP), and its major metabolite, DEP, in rats. Levels of serum lipids and, sex hormones were measured using immunoassay kits. Gut hormones and inflammatory cytokines were assessed using a multiplexing kit, and the gut microbiota was evaluated by 16S rRNA gene sequencing. After a 24-week exposure period, both TAP and DEP significantly decreased serum levels of triglycerides, cholesterol, low-density lipoprotein cholesterol, and IL-6 (p < 0.05). However, DEP exposure had a stronger effect on serum estradiol (p < 0.05) than TAP, whereas only TAP inhibited the secretion of gut hormones. Both TAP and DEP enriched the pathogenic genera Oscillibacter, Peptococcus and Paraprevotella in the gut, and TAP also enriched enteritis-related genera Roseburia and Oscillibacter, which may affect the secretion of gut hormones. These findings indicate that the use of dialkyl phosphates as markers of OPs to examine the correlations of OP exposure with diseases may only provide partial information, especially for diseases related to gut health and the endocrine system.
Collapse
Affiliation(s)
- Fang-Wei Yang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Bing Fang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Guo-Fang Pang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Ming Zhang
- School of Food Science and Chemical Engineering, Beijing Technology and Business University, Beijing, China
| | - Fa-Zheng Ren
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Nutrition and Health, China Agricultural University, Beijing, China
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, and Beijing Laboratory of Food Quality and Safety, China Agricultural University, Beijing, China
| |
Collapse
|
13
|
Tirpák F, Greifová H, Lukáč N, Stawarz R, Massányi P. Exogenous Factors Affecting the Functional Integrity of Male Reproduction. Life (Basel) 2021; 11:213. [PMID: 33803103 PMCID: PMC8001766 DOI: 10.3390/life11030213] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/04/2021] [Accepted: 03/07/2021] [Indexed: 12/16/2022] Open
Abstract
Natural processes along with increased industrial production and the irresponsible behavior of mankind have resulted in environmental pollution. Environmental pollutants can be categorized based on their characteristics and appearance into the following groups: physical, biological, and chemical. Every single one of them represents a serious threat to the male reproductive tract despite the different modes of action. Male gonads and gametes are especially vulnerable to the effect of exogenous factors; therefore, they are considered a reliable indicator of environmental pollution. The impact of xenobiotics or radiation leads to an irreversible impairment of fertility displayed by histological changes, modulated androgen production, or compromised spermatozoa (or germ cells) quality. The present article reviews the exogenous threats, male reproductive system, the mode of action, and overall impact on the reproductive health of humans and animals.
Collapse
Affiliation(s)
- Filip Tirpák
- AgroBioTech Research Centre, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovakia
| | - Hana Greifová
- Department of Animal Physiology, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovakia; (H.G.); (N.L.)
| | - Norbert Lukáč
- Department of Animal Physiology, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovakia; (H.G.); (N.L.)
| | - Robert Stawarz
- Institute of Biology, Pedagogical University of Krakow, Podchorazych 2, 30-084 Krakow, Poland;
| | - Peter Massányi
- Department of Animal Physiology, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovakia; (H.G.); (N.L.)
- Institute of Biology, Pedagogical University of Krakow, Podchorazych 2, 30-084 Krakow, Poland;
| |
Collapse
|
14
|
Hess RA, Sharpe RM, Hinton BT. Estrogens and development of the rete testis, efferent ductules, epididymis and vas deferens. Differentiation 2021; 118:41-71. [PMID: 33441255 PMCID: PMC8026493 DOI: 10.1016/j.diff.2020.11.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 11/29/2020] [Indexed: 02/07/2023]
Abstract
Estrogen has always been considered the female hormone and testosterone the male hormone. However, estrogen's presence in the testis and deleterious effects of estrogen treatment during development have been known for nearly 90 years, long before estrogen receptors (ESRs) were discovered. Eventually it was learned that testes actually synthesize high levels of estradiol (E2) and sequester high concentrations in the reproductive tract lumen, which seems contradictory to the overwhelming number of studies showing reproductive pathology following exogenous estrogen exposures. For too long, the developmental pathology of estrogen has dominated our thinking, even resulting in the "estrogen hypothesis" as related to the testicular dysgenesis syndrome. However, these early studies and the development of an Esr1 knockout mouse led to a deluge of research into estrogen's potential role in and disruption of development and function of the male reproductive system. What is new is that estrogen action in the male cannot be divorced from that of androgen. This paper presents what is known about components of the estrogen pathway, including its synthesis and target receptors, and the need to achieve a balance between androgen- and estrogen-action in male reproductive tract differentiation and adult functions. The review focuses on what is known regarding development of the male reproductive tract, from the rete testis to the vas deferens, and examines the expression of estrogen receptors and presence of aromatase in the male reproductive system, traces the evidence provided by estrogen-associated knockout and transgenic animal models and discusses the effects of fetal and postnatal exposures to estrogens. Hopefully, there will be enough here to stimulate discussions and new investigations of the androgen:estrogen balance that seems to be essential for development of the male reproductive tract.
Collapse
Affiliation(s)
- Rex A Hess
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois Urbana-Champaign, IL, 61802 USA and Epivara, Inc., Research Park, 60 Hazelwood Dr., Suite 230G, Champaign, IL, 61820, USA.
| | - Richard M Sharpe
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
| | - Barry T Hinton
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, USA.
| |
Collapse
|
15
|
Aprea I, Nöthe-Menchen T, Dougherty GW, Raidt J, Loges NT, Kaiser T, Wallmeier J, Olbrich H, Strünker T, Kliesch S, Pennekamp P, Omran H. Motility of efferent duct cilia aids passage of sperm cells through the male reproductive system. Mol Hum Reprod 2021; 27:gaab009. [PMID: 33561200 PMCID: PMC7936721 DOI: 10.1093/molehr/gaab009] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 01/06/2021] [Indexed: 02/06/2023] Open
Abstract
Motile cilia line the efferent ducts of the mammalian male reproductive tract. Several recent mouse studies have demonstrated that a reduced generation of multiple motile cilia in efferent ducts is associated with obstructive oligozoospermia and fertility issues. However, the sole impact of efferent duct cilia dysmotility on male infertility has not been studied so far either in mice or human. Using video microscopy, histological- and ultrastructural analyses, we examined male reproductive tracts of mice deficient for the axonemal motor protein DNAH5: this defect exclusively disrupts the outer dynein arm (ODA) composition of motile cilia but not the ODA composition and motility of sperm flagella. These mice have immotile efferent duct cilia that lack ODAs, which are essential for ciliary beat generation. Furthermore, they show accumulation of sperm in the efferent duct. Notably, the ultrastructure and motility of sperm from these males are unaffected. Likewise, human individuals with loss-of-function DNAH5 mutations present with reduced sperm count in the ejaculate (oligozoospermia) and dilatations of the epididymal head but normal sperm motility, similar to DNAH5 deficient mice. The findings of this translational study demonstrate, in both mice and men, that efferent duct ciliary motility is important for male reproductive fitness and uncovers a novel pathomechanism distinct from primary defects of sperm motility (asthenozoospermia). If future work can identify environmental factors or defects in genes other than DNAH5 that cause efferent duct cilia dysmotility, this will help unravel other causes of oligozoospermia and may influence future practices in genetic and fertility counseling as well as ART.
Collapse
Affiliation(s)
- Isabella Aprea
- Department of General Pediatrics, University Hospital Muenster, Muenster 48149, Germany
| | - Tabea Nöthe-Menchen
- Department of General Pediatrics, University Hospital Muenster, Muenster 48149, Germany
| | - Gerard W Dougherty
- Department of General Pediatrics, University Hospital Muenster, Muenster 48149, Germany
| | - Johanna Raidt
- Department of General Pediatrics, University Hospital Muenster, Muenster 48149, Germany
| | - Niki T Loges
- Department of General Pediatrics, University Hospital Muenster, Muenster 48149, Germany
| | - Thomas Kaiser
- Department of General Pediatrics, University Hospital Muenster, Muenster 48149, Germany
| | - Julia Wallmeier
- Department of General Pediatrics, University Hospital Muenster, Muenster 48149, Germany
| | - Heike Olbrich
- Department of General Pediatrics, University Hospital Muenster, Muenster 48149, Germany
| | - Timo Strünker
- Department of Clinical Andrology, Centre of Reproductive Medicine and Andrology, University Hospital Muenster, Muenster 48149, Germany
| | - Sabine Kliesch
- Department of Clinical Andrology, Centre of Reproductive Medicine and Andrology, University Hospital Muenster, Muenster 48149, Germany
| | - Petra Pennekamp
- Department of General Pediatrics, University Hospital Muenster, Muenster 48149, Germany
| | - Heymut Omran
- Department of General Pediatrics, University Hospital Muenster, Muenster 48149, Germany
| |
Collapse
|
16
|
Hess RA, Cooke PS. Estrogen in the male: a historical perspective. Biol Reprod 2019; 99:27-44. [PMID: 29438493 PMCID: PMC6044326 DOI: 10.1093/biolre/ioy043] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 02/08/2018] [Indexed: 12/18/2022] Open
Abstract
Estrogens have traditionally been considered female hormones. Nevertheless, the presence of estrogen in males has been known for over 90 years. Initial studies suggested that estrogen was deleterious to male reproduction because exogenous treatments induced developmental abnormalities. However, demonstrations of estrogen synthesis in the testis and high concentrations of 17β-estradiol in rete testis fluid suggested that the female hormone might have a function in normal male reproduction. Identification of estrogen receptors and development of biological radioisotope methods to assess estradiol binding revealed that the male reproductive tract expresses estrogen receptor extensively from the neonatal period to adulthood. This indicated a role for estrogens in normal development, especially in efferent ductules, whose epithelium is the first in the male reproductive tract to express estrogen receptor during development and a site of exceedingly high expression. In the 1990s, a paradigm shift occurred in our understanding of estrogen function in the male, ushered in by knockout mouse models where estrogen production or expression of its receptors was not present. These knockout animals revealed that estrogen's main receptor (estrogen receptor 1 [ESR1]) is essential for male fertility and development of efferent ductules, epididymis, and prostate, and that loss of only the membrane fraction of ESR1 was sufficient to induce extensive male reproductive abnormalities and infertility. This review provides perspectives on the major discoveries and developments that led to our current knowledge of estrogen's importance in the male reproductive tract and shaped our evolving concept of estrogen's physiological role in the male.
Collapse
Affiliation(s)
- Rex A Hess
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Paul S Cooke
- Department of Physiological Sciences, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
17
|
Zhai J, Geng X, Ding T, Li J, Tang J, Chen D, Cui L, Wang Q. An increase of estrogen receptor α protein level regulates BDE-209-mediated blood-testis barrier disruption during spermatogenesis in F1 mice. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:4801-4820. [PMID: 30565106 DOI: 10.1007/s11356-018-3784-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 11/15/2018] [Indexed: 06/09/2023]
Abstract
Deca-bromodiphenyl ether (BDE-209) regulates various aspects of spermatogenesis and male fertility through its effect on estrogen receptor α (ERα), but the underlying mechanism remains unclear. Because molecular mechanisms such as remodeling of the blood-testis barrier (BTB) play crucial roles in spermatogenesis, we investigated the disruptive effects of ERα agonists on the BTB in spermatogenesis. In this study, 0, 300, and 500 mg/kg/day of BDE-209 were administered to pregnant adult mice by oral gavage from gestation day 7 to postnatal day 21. SerW3 cells were treated with methylpiperidino pyrazole (MPP) for 30 min before being treated with 50 μg/mL of BDE-209. BDE-209 increases ERα in time- and dose-dependent manners and decreases formin 1 and BTB-associated protein in F1 male mice. Furthermore, BDE-209 impairs the structure and function of the BTB. Activation of ERα signaling could disrupt the BTB, leading to spermatogenesis dysfunction. The results identified the role of ERα in BTB disruption during spermatogenesis and suggested that BTB disruption occurs because of exposure to BDE-209, which could potentially affect spermatogenesis. In conclusion, Sertoli cells seem to be the primary target of BDE-209 in the perinatal period, and this period constitutes a critical window of susceptibility to BDE-209. Also, the SerW3 cell model may not be a particularly useful cell model for studying the function of the cytoskeleton.
Collapse
Affiliation(s)
- Jinxia Zhai
- Department of Occupational and Environmental Health, School of Public Health, Anhui Medical University, Meishan Rd 81, Hefei, 230032, China.
| | - Xiya Geng
- Department of Occupational and Environmental Health, School of Public Health, Anhui Medical University, Meishan Rd 81, Hefei, 230032, China
| | - Tao Ding
- Department of Occupational and Environmental Health, School of Public Health, Anhui Medical University, Meishan Rd 81, Hefei, 230032, China
| | - Jun Li
- Department of Occupational and Environmental Health, School of Public Health, Anhui Medical University, Meishan Rd 81, Hefei, 230032, China
| | - Jing Tang
- Department of Occupational and Environmental Health, School of Public Health, Anhui Medical University, Meishan Rd 81, Hefei, 230032, China
| | - Daojun Chen
- Department of Occupational and Environmental Health, School of Public Health, Anhui Medical University, Meishan Rd 81, Hefei, 230032, China
| | - Longjiang Cui
- Department of Occupational and Environmental Health, School of Public Health, Anhui Medical University, Meishan Rd 81, Hefei, 230032, China
| | - Qizhi Wang
- School of Energy and Environment, Southeast University, Sipailou Rd 2, Nanjing, 210018, China
| |
Collapse
|
18
|
Motile cilia of the male reproductive system require miR-34/miR-449 for development and function to generate luminal turbulence. Proc Natl Acad Sci U S A 2019; 116:3584-3593. [PMID: 30659149 DOI: 10.1073/pnas.1817018116] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Cilia are cell-surface, microtubule-based organelles that project into extracellular space. Motile cilia are conserved throughout eukaryotes, and their beat induces the flow of fluid, relative to cell surfaces. In mammals, the coordinated beat of motile cilia provides highly specialized functions associated with the movement of luminal contents, as seen with metachronal waves transporting mucus in the respiratory tract. Motile cilia are also present in the male and female reproductive tracts. In the female, wave-like motions of oviductal cilia transport oocytes and embryos toward the uterus. A similar function has been assumed for motile cilia in efferent ductules of the male-i.e., to transport immotile sperm from rete testis into the epididymis. However, we report here that efferent ductal cilia in the male do not display a uniform wave-like beat to transport sperm solely in one direction, but rather exert a centripetal force on luminal fluids through whip-like beating with continual changes in direction, generating turbulence, which maintains immotile spermatozoa in suspension within the lumen. Genetic ablation of two miRNA clusters (miR-34b/c and -449a/b/c) led to failure in multiciliogenesis in murine efferent ductules due to dysregulation of numerous genes, and this mouse model allowed us to demonstrate that loss of efferent duct motile cilia causes sperm aggregation and agglutination, luminal obstruction, and sperm granulomas, which, in turn, induce back-pressure atrophy of the testis and ultimately male infertility.
Collapse
|
19
|
Leite GAA, de Barros JWF, Martins ADC, Anselmo-Franci JA, Barbosa F, Kempinas WDG. Ascorbic acid supplementation ameliorates testicular hormonal signaling, sperm production and oxidative stress in male rats exposed to rosuvastatin during pre-puberty. J Appl Toxicol 2018; 39:305-321. [DOI: 10.1002/jat.3720] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 07/25/2018] [Accepted: 08/03/2018] [Indexed: 01/22/2023]
Affiliation(s)
- Gabriel Adan Araujo Leite
- Graduate Program in Cell and Structural Biology, Institute of Biology; State University of Campinas - UNICAMP; Campinas Brazil
- Department of Morphology, São Paulo State University (Unesp); Institute of Biosciences; Botucatu Brazil
| | | | - Airton da Cunha Martins
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto; University of São Paulo - USP; Ribeirão Preto Brazil
| | - Janete Aparecida Anselmo-Franci
- Department of Morphology, Physiology and Basic Pathology, School of Dentistry of Ribeirão Preto; USP-University of São Paulo, Ribeirão Preto; São Paulo Brazil
| | - Fernando Barbosa
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto; University of São Paulo - USP; Ribeirão Preto Brazil
| | - Wilma De Grava Kempinas
- Department of Morphology, São Paulo State University (Unesp); Institute of Biosciences; Botucatu Brazil
| |
Collapse
|
20
|
Curley M, Milne L, Smith S, Atanassova N, Rebourcet D, Darbey A, Hadoke PWF, Wells S, Smith LB. Leukemia Inhibitory Factor-Receptor is Dispensable for Prenatal Testis Development but is Required in Sertoli cells for Normal Spermatogenesis in Mice. Sci Rep 2018; 8:11532. [PMID: 30068994 PMCID: PMC6070476 DOI: 10.1038/s41598-018-30011-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 07/19/2018] [Indexed: 12/14/2022] Open
Abstract
Leukemia inhibitory factor (LIF), a pleiotropic cytokine belonging to the interleukin-6 family, is most often noted for its role in maintaining the balance between stem cell proliferation and differentiation. In rodents, LIF is expressed in both the fetal and adult testis; with the peritubular myoid (PTM) cells thought to be the main site of production. Given their anatomical location, LIF produced by PTM cells may act both on intratubular and interstitial cells to influence spermatogenesis and steroidogenesis respectively. Indeed, the leukemia inhibitory factor receptor (LIFR) is expressed in germ cells, Sertoli cells, Leydig cells, PTM cells and testicular macrophages, suggesting that LIF signalling via LIFR may be a key paracrine regulator of testicular function. However, a precise role(s) for testicular LIFR-signalling in vivo has not been established. To this end, we generated and characterised the testicular phenotype of mice lacking LIFR either in germ cells, Sertoli cells or both, to identify a role for LIFR-signalling in testicular development/function. Our analyses reveal that LIFR is dispensable in germ cells for normal spermatogenesis. However, Sertoli cell LIFR ablation results in a degenerative phenotype, characterised by abnormal germ cell loss, sperm stasis, seminiferous tubule distention and subsequent atrophy of the seminiferous tubules.
Collapse
Affiliation(s)
- Michael Curley
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, United Kingdom
| | - Laura Milne
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, United Kingdom
| | - Sarah Smith
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, United Kingdom
| | - Nina Atanassova
- Institute of Experimental Morphology, Pathology and Anthropology with Museum, Bulgarian Academy of Sciences, 1113, Sofia, Bulgaria
| | - Diane Rebourcet
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, United Kingdom
| | - Annalucia Darbey
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, United Kingdom
| | - Patrick W F Hadoke
- The British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, EH16 4TJ, United Kingdom
| | - Sara Wells
- Mary Lyons Centre, MRC Harwell, Harwell Campus, Oxfordshire, OX11 ORD, United Kingdom
| | - Lee B Smith
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, United Kingdom. .,School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW, 2308, Australia.
| |
Collapse
|
21
|
Cooke PS, Nanjappa MK, Ko C, Prins GS, Hess RA. Estrogens in Male Physiology. Physiol Rev 2017; 97:995-1043. [PMID: 28539434 PMCID: PMC6151497 DOI: 10.1152/physrev.00018.2016] [Citation(s) in RCA: 283] [Impact Index Per Article: 40.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 01/06/2017] [Accepted: 01/17/2017] [Indexed: 02/06/2023] Open
Abstract
Estrogens have historically been associated with female reproduction, but work over the last two decades established that estrogens and their main nuclear receptors (ESR1 and ESR2) and G protein-coupled estrogen receptor (GPER) also regulate male reproductive and nonreproductive organs. 17β-Estradiol (E2) is measureable in blood of men and males of other species, but in rete testis fluids, E2 reaches concentrations normally found only in females and in some species nanomolar concentrations of estrone sulfate are found in semen. Aromatase, which converts androgens to estrogens, is expressed in Leydig cells, seminiferous epithelium, and other male organs. Early studies showed E2 binding in numerous male tissues, and ESR1 and ESR2 each show unique distributions and actions in males. Exogenous estrogen treatment produced male reproductive pathologies in laboratory animals and men, especially during development, and studies with transgenic mice with compromised estrogen signaling demonstrated an E2 role in normal male physiology. Efferent ductules and epididymal functions are dependent on estrogen signaling through ESR1, whose loss impaired ion transport and water reabsorption, resulting in abnormal sperm. Loss of ESR1 or aromatase also produces effects on nonreproductive targets such as brain, adipose, skeletal muscle, bone, cardiovascular, and immune tissues. Expression of GPER is extensive in male tracts, suggesting a possible role for E2 signaling through this receptor in male reproduction. Recent evidence also indicates that membrane ESR1 has critical roles in male reproduction. Thus estrogens are important physiological regulators in males, and future studies may reveal additional roles for estrogen signaling in various target tissues.
Collapse
Affiliation(s)
- Paul S Cooke
- Department of Physiological Sciences, University of Florida, Gainesville, Florida; Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, Illinois; Department of Urology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Manjunatha K Nanjappa
- Department of Physiological Sciences, University of Florida, Gainesville, Florida; Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, Illinois; Department of Urology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - CheMyong Ko
- Department of Physiological Sciences, University of Florida, Gainesville, Florida; Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, Illinois; Department of Urology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Gail S Prins
- Department of Physiological Sciences, University of Florida, Gainesville, Florida; Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, Illinois; Department of Urology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Rex A Hess
- Department of Physiological Sciences, University of Florida, Gainesville, Florida; Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, Illinois; Department of Urology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
22
|
Nanjappa MK, Mesa AM, Tevosian SG, de Armas L, Hess RA, Bagchi IC, Cooke PS. Membrane estrogen receptor 1 is required for normal reproduction in male and female mice. JOURNAL OF ENDOCRINOLOGY AND REPRODUCTION : JER 2017; 21:1-14. [PMID: 34321782 PMCID: PMC8315114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Steroid hormones, acting through their cognate nuclear receptors, are critical for many reproductive and non-reproductive functions. Over the past two decades, it has become increasingly clear that in addition to cytoplasmic/nuclear steroid receptors that alter gene transcription when liganded, a small fraction of cellular steroid receptors are localized to the cell membranes, where they mediate rapid steroid hormone effects. 17β-Estradiol (E2), a key steroid hormone for both male and female reproduction, acts predominately through its main receptor, estrogen receptor 1 (ESR1). Most ESR1 is nuclear; however, 5-10% of ESR1 is localized to the cell membrane after being palmitoylated at cysteine 451 in mice. This review discusses reproductive phenotypes of a newly-developed mouse model with a C451A point mutation that precludes membrane targeting of ESR1. This transgenic mouse, termed the nuclear-only ESR1 (NOER) mouse, shows extensive male and female reproductive abnormalities and infertility despite normally functional nuclear ESR1 (nESR1). These results provide the first in vivo evidence that membrane-initiated E2/ESR1 signaling is required for normal male and female reproductive functions and fertility. Signaling mechanisms for membrane ESR1 (mESR1), as well as how mESR1 works with nESR1 to mediate estrogen effects, are still being established. We discuss some possible mechanisms by which mESR1 might facilitate nESR1 signaling, as well as the emerging evidence that mESR1 might be a major mediator of epigenetic effects of estrogens, which are potentially linked to various adult-onset pathologies.
Collapse
Affiliation(s)
| | - Ana M. Mesa
- Department of Physiological Sciences, University of Florida, Gainesville, FL 32610, USA
| | - Sergei G. Tevosian
- Department of Physiological Sciences, University of Florida, Gainesville, FL 32610, USA
| | - Laura de Armas
- Department of Physiological Sciences, University of Florida, Gainesville, FL 32610, USA
| | - Rex A. Hess
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Indrani C. Bagchi
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Paul S. Cooke
- Department of Physiological Sciences, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
23
|
Wang YY, Lin YH, Wu YN, Chen YL, Lin YC, Cheng CY, Chiang HS. Loss of SLC9A3 decreases CFTR protein and causes obstructed azoospermia in mice. PLoS Genet 2017; 13:e1006715. [PMID: 28384194 PMCID: PMC5398719 DOI: 10.1371/journal.pgen.1006715] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 04/20/2017] [Accepted: 03/24/2017] [Indexed: 01/21/2023] Open
Abstract
Mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene cause cystic fibrosis (CF) and are associated with congenital bilateral absence of the vas deferens (CBAVD), which is the major cause of infertility in male patients with CF. However, most Taiwanese patients with CBAVD do not carry major CFTR mutations. Some patients have a single copy deletion of the solute carrier family 9 isoform 3 (SLC9A3) gene. SLC9A3 is a Na+/H+ exchanger, and depleted Slc9a3 in male mice causes infertility due to the abnormal dilated lumen of the rete testis and efferent ductules. Furthermore, SLC9A3 interacts with CFTR in the pancreatic duct and functions as a genetic modifier of CF. However, SLC9A3 function and its relation to CFTR expression in the male reproductive tract in vivo remain elusive. In the present study, we found that CFTR expression was dramatically decreased in the epididymis and vas deferens of Slc9a3 knockout mice. Adult Slc9a3-/- mice showed not only significantly decreased epididymis and vas deferens weight but also increased testis weight. Furthermore, Slc9a3-/- mice developed obstructive azoospermia because of abnormal abundant secretions and calcification in the lumen of the reproductive tract. Ultrastructural analysis of the epithelium in Slc9a3–/–epididymis and vas deferens displayed disorganized and reduced number of stereocilia and numerous secretory apparatuses. Our data revealed that interdependence between SLC9A3 and CFTR is critical for maintaining a precise microenvironment in the epithelial cytoarchitecture of the male reproductive tract. The Slc9a3-deficient mice with impaired male excurrent ducts in this study provide proof for our clinical findings that some Taiwanese of CBAVD carry SLC9A3 deletion but without major CFTR mutations. Cystic fibrosis (CF) is the most common inherited life-threatening disease in Caucasians. The most well-known cause of CF is a genetic defect in CFTR, an apical membrane chloride and bicarbonate channel. The symptoms of CF include defects in the respiratory, digestive, and male reproductive systems. Most male patients with CF are infertile due to congenital bilateral absence of the vas deferens (CBAVD), which leads to obstructive azoospermia. Nevertheless, Taiwanese patients with CBAVD do not carry the common mutations of CFTR found in Caucasians. We have identified a potential candidate, SLC9A3, of which a single copy is lost in Taiwanese patients with CBAVD. In addition to the previously reported role of SLC9A3 in the digestive system and efferent ductules, we now report that the SLC9A3 deficiency causes obstructive azoospermia and impairs the epithelial structure of the reproductive tract. Loss of SLC9A3 also leads to dramatic reduced expression of CFTR in the reproductive tract. We suggest that the interplay between SLC9A3 and CFTR is responsible for CF-related infertility. Thus, we have characterized a potential critical player in the pathogenesis of CBAVD and provide a new diagnostic candidate for Asian patients with CBAVD.
Collapse
Affiliation(s)
- Ya-Yun Wang
- Department of Chemistry, Fu Jen Catholic University, New Taipei City, Taiwan
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Ying-Hung Lin
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Yi-No Wu
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Yen-Lin Chen
- Department of Chemistry, Fu Jen Catholic University, New Taipei City, Taiwan
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City, Taiwan
- Department of Pathology, Cardinal Tien Hospital, New Taipei City, Taiwan
| | - Yung-Chih Lin
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Chiao-Yin Cheng
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Han-Sun Chiang
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City, Taiwan
- Department of Urology, Taipei Medical University Hospital, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
24
|
Danielian PS, Hess RA, Lees JA. E2f4 and E2f5 are essential for the development of the male reproductive system. Cell Cycle 2016; 15:250-60. [PMID: 26825228 DOI: 10.1080/15384101.2015.1121350] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The E2F transcription factors are primarily implicated in the regulation of entry and exit from the cell cycle. However, in vivo studies have established additional roles for E2Fs during organ development and homeostasis. With the goal of addressing the intestinal requirements of E2f4 and E2f5, we crossed mice carrying Vil-cre, E2f4 conditional and E2f5 germline alleles. E2f4 deletion had no detectable effect on intestinal development. However, E2f4f/f;E2f5+/-;Vil-cre males, but not E2f4f/f;Vil-cre littermates, were unexpectedly sterile. This defect was not due to defective spermatogenesis. Instead, the seminiferous tubules and rete testes showed significant dilation, and spermatozoa accumulated aberrantly in the rete testis and efferent ducts. Our data show that these problems result from defective efferent ducts, a tissue whose primary function is to concentrate sperm through fluid absorption. First, Vil-cre expression, and consequent E2F4 loss, was specific to the efferent ducts and not other reproductive tract tissues. Second, the E2f4f/f;E2f5+/-;Vil-cre efferent ducts had completely lost multiciliated cells and greatly reduced levels of critical absorptive cell proteins: aquaporin1, a water channel protein, and clusterin, an endocytic marker. Collectively, the observed testis phenotypes suggest a fluid flux defect. Remarkably, we observed rete testis dilation prior to the normal time of seminiferous fluid production, arguing that the efferent duct defects promote excessive secretory activity within the reproductive tract. Finally, we also detect key aspects of these testis defects in E2f5-/- mice. Thus, we conclude that E2f4 and E2f5 display overlapping roles in controlling the normal development of the male reproductive system.
Collapse
Affiliation(s)
- Paul S Danielian
- a David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge , MA , USA
| | - Rex A Hess
- b Reproductive Biology & Toxicology , Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois , Urbana , IL , USA
| | - Jacqueline A Lees
- a David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge , MA , USA
| |
Collapse
|
25
|
Sipilä P, Björkgren I. Segment-specific regulation of epididymal gene expression. Reproduction 2016; 152:R91-9. [DOI: 10.1530/rep-15-0533] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 05/23/2016] [Indexed: 01/24/2023]
Abstract
The epididymis is necessary for post-testicular sperm maturation. During their epididymal transit, spermatozoa gain ability for progressive movement and fertilization. The epididymis is composed of several segments that have distinct gene expression profiles that enable the establishment of the changing luminal environment required for sperm maturation. The epididymal gene expression is regulated by endocrine, lumicrine, and paracrine factors in a segment-specific manner. Thus, in addition to its importance for male fertility, the epididymis is a valuable model tissue for studying the regulation of gene expression. This review concentrates on recent advances in understanding the androgen, small RNA, and epigenetically mediated regulation of segment-specific gene expression in the epididymis.
Collapse
|
26
|
Nanjappa MK, Hess RA, Medrano TI, Locker SH, Levin ER, Cooke PS. Membrane-Localized Estrogen Receptor 1 Is Required for Normal Male Reproductive Development and Function in Mice. Endocrinology 2016; 157:2909-19. [PMID: 27145009 PMCID: PMC4929544 DOI: 10.1210/en.2016-1085] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Estrogen receptor 1 (ESR1) mediates major reproductive functions of 17β-estradiol (E2). Male Esr1 knockout (Esr1KO) mice are infertile due to efferent ductule and epididymal abnormalities. The majority of ESR1 is nuclear/cytoplasmic; however, a small fraction is palmitoylated at cysteine 451 in mice and localized to cell membranes, in which it mediates rapid E2 actions. This study used an Esr1 knock-in mouse containing an altered palmitoylation site (C451A) in ESR1 that prevented cell membrane localization, although nuclear ESR1 was expressed. These nuclear-only estrogen receptor 1 (NOER) mice were used to determine the roles of membrane ESR1 in males. Epididymal sperm motility was reduced 85% in 8-month-old NOER mice compared with wild-type controls. The NOER mice had decreased epididymal sperm viability and greater than 95% of sperm had abnormalities, including coiled midpieces and tails, absent heads, and folded tails; this was comparable to 4-month Esr1KO males. At 8 months, daily sperm production in NOER males was reduced 62% compared with controls. The NOER mice had histological changes in the rete testes, efferent ductules, and seminiferous tubules that were comparable with those previously observed in Esr1KO males. Serum T was increased in NOER males, but FSH, LH, and E2 were unchanged. Critically, NOER males were initially subfertile, becoming infertile with advancing age. These findings identify a previously unknown role for membrane ESR1 in the development of normal sperm and providing an adequate environment for spermatogenesis.
Collapse
Affiliation(s)
- Manjunatha K Nanjappa
- Department of Physiological Sciences (M.K.N., T.I.M., S.H.L., P.S.C.), University of Florida, Gainesville, Florida 32610; Department of Comparative Biosciences (R.A.H.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61801; Division of Endocrinology (E.R.L.), Department of Medicine, University of California, Irvine, Irvine, California 92697; and Department of Veterans Affairs Medical Center (E.R.L.), Long Beach, Long Beach, California 90822
| | - Rex A Hess
- Department of Physiological Sciences (M.K.N., T.I.M., S.H.L., P.S.C.), University of Florida, Gainesville, Florida 32610; Department of Comparative Biosciences (R.A.H.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61801; Division of Endocrinology (E.R.L.), Department of Medicine, University of California, Irvine, Irvine, California 92697; and Department of Veterans Affairs Medical Center (E.R.L.), Long Beach, Long Beach, California 90822
| | - Theresa I Medrano
- Department of Physiological Sciences (M.K.N., T.I.M., S.H.L., P.S.C.), University of Florida, Gainesville, Florida 32610; Department of Comparative Biosciences (R.A.H.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61801; Division of Endocrinology (E.R.L.), Department of Medicine, University of California, Irvine, Irvine, California 92697; and Department of Veterans Affairs Medical Center (E.R.L.), Long Beach, Long Beach, California 90822
| | - Seth H Locker
- Department of Physiological Sciences (M.K.N., T.I.M., S.H.L., P.S.C.), University of Florida, Gainesville, Florida 32610; Department of Comparative Biosciences (R.A.H.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61801; Division of Endocrinology (E.R.L.), Department of Medicine, University of California, Irvine, Irvine, California 92697; and Department of Veterans Affairs Medical Center (E.R.L.), Long Beach, Long Beach, California 90822
| | - Ellis R Levin
- Department of Physiological Sciences (M.K.N., T.I.M., S.H.L., P.S.C.), University of Florida, Gainesville, Florida 32610; Department of Comparative Biosciences (R.A.H.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61801; Division of Endocrinology (E.R.L.), Department of Medicine, University of California, Irvine, Irvine, California 92697; and Department of Veterans Affairs Medical Center (E.R.L.), Long Beach, Long Beach, California 90822
| | - Paul S Cooke
- Department of Physiological Sciences (M.K.N., T.I.M., S.H.L., P.S.C.), University of Florida, Gainesville, Florida 32610; Department of Comparative Biosciences (R.A.H.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61801; Division of Endocrinology (E.R.L.), Department of Medicine, University of California, Irvine, Irvine, California 92697; and Department of Veterans Affairs Medical Center (E.R.L.), Long Beach, Long Beach, California 90822
| |
Collapse
|