1
|
Chahdi A, Jorgez C, Rosenfeld JA, Neetu N, Seth A. Androgen receptor ubiquitination links KCTD13 to genitourinary tract defects. FASEB J 2025; 39:e70406. [PMID: 39968753 DOI: 10.1096/fj.202402072rr] [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: 09/03/2024] [Revised: 01/09/2025] [Accepted: 02/10/2025] [Indexed: 02/20/2025]
Abstract
The potassium channel tetramerization domain containing 13 (KCTD13) protein is a substrate-specific adapter for cullin3-based E3 ubiquitin ligase. Patients with copy number variants at this locus exhibit genitourinary tract anomalies. In this study, we show that decreased androgen receptor (AR) protein level correlated with increased AR ubiquitination in the testis of Kctd13-deficient mice, suggesting that KCTD13 inhibits AR ubiquitination. KCTD13 increased CUL3-dependent AR ubiquitination but had no effect on CUL3 binding to AR, confirming the role of KCTD13 as an adaptor of CUL3 ligase. Recombinant KCTD13 directly binds to recombinant AR, and the BTB domain of KCTD13 is critical for binding both the N-terminal domain of AR and STUB1. Moreover, KCTD13 dose-dependently decreased STUB1 binding to AR resulting in decreased AR ubiquitination. KCTD13 ΔBTB was unable to bind to AR and subsequently failed to block STUB1-mediated AR ubiquitination, strongly suggesting that reduced AR ubiquitination is dependent on KCTD13 ability to dissociate AR/STUB1 complex. Furthermore, KCTD13 increased the expression of AR target gene, FOXJ1, whereas KCTD13 ΔBTB had no effect. Our data reveal a distinctive mode of action of KCTD13 on AR ubiquitination depending on the E3 ubiquitin ligase involved: (1) KCTD13 increased CUL3-dependent AR ubiquitination but had no effect on CUL3 binding to AR; and (2) KCTD13 decreased STUB1-mediated AR ubiquitination by decreasing STUB1 binding to AR thus preventing AR ubiquitination. We hypothesize that in the testes of Kctd13-deficient mice, the absence of KCTD13 results in increased binding of STUB1 to AR leading to increased AR ubiquitination and degradation.
Collapse
Affiliation(s)
- Ahmed Chahdi
- Department of Surgery, Nemours Children's Health, Orlando, Florida, USA
- College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Carolina Jorgez
- Scott Department of Urology, Baylor College of Medicine, Houston, Texas, USA
| | - Jill A Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Neetu Neetu
- Department of Biochemistry, Baylor College of Medicine, Houston, Texas, USA
| | - Abhishek Seth
- Department of Surgery, Nemours Children's Health, Orlando, Florida, USA
- College of Medicine, University of Central Florida, Orlando, Florida, USA
| |
Collapse
|
2
|
Silveira JM, Cesar Dos Santos A, Calado de Brito DC, de Oliveira MF, Conley AJ, de Assis Neto AC. Morphohistometric and steroidogenic parameters during testicular and epididymal differentiation in cavy (Galea spixii) fetuses. Reprod Biol 2024; 24:100829. [PMID: 38039944 DOI: 10.1016/j.repbio.2023.100829] [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: 04/24/2023] [Revised: 10/30/2023] [Accepted: 11/17/2023] [Indexed: 12/03/2023]
Abstract
Sexual differentiation and steroidogenic mechanisms have an important impact on postnatal gonadal phenotypic development. Thus, establishing the activities that lead to male phenotypic development can provide a better understanding of this process. This study examined the prenatal development of cavies to establish morphological and histometric development patterns and protein and enzyme immunolocalization processes that are responsible for androgen synthesis in the testes and epididymis. Histological and histometric analyses of the diameter of the seminiferous cords and epididymal ducts of male fetuses on Days 25, 30, 40, and 50 were performed, as well as immunohistochemistry of the steroidogenic enzymes 5α-reductase and 17β-HSD, the androgen receptor, and the anti-Müllerian hormone (AMH). Our findings showed a cellular grouping of gonocytes from Day 30 onward that was characteristic of the seminiferous cord, which was not present in the lumen at any of the studied dates. From Day 50 onward, the differentiation of the three anatomical regions of the epididymis was evident, the head (caput), body (corpus), and tail (cauda), with tissue distinctions. Furthermore, the diameters of the seminiferous cords and epididymal ducts significantly increased with age. On Day 50, the tail showed the greatest diameter of the three regions. The Sertoli and Leydig cells exhibited AMH immunoreactivity at all dates. In addition, the Leydig cells and epididymal epithelial tissue were immunopositive for 5α-reductase, 17β-HSD, and the androgen receptor; therefore, these factors influenced the development and maintenance of the testis and epididymis during cavy prenatal development.
Collapse
Affiliation(s)
- Júlia Moreira Silveira
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Amilton Cesar Dos Santos
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | | | | | - Alan James Conley
- Department of Population Health & Reproduction, School of Veterinary Medicine, University of California, UC, Davis, CA, USA
| | - Antonio Chaves de Assis Neto
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
3
|
Lemos GAA, Santos AC, Brito DCC, Novaes MAS, Assis Neto AC. Steroidogenic activity and morphological characterization of prenatal testes and epididymis of guinea pig (Cavia porcellus). Anim Reprod Sci 2024; 261:107407. [PMID: 38217925 DOI: 10.1016/j.anireprosci.2023.107407] [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: 08/20/2023] [Accepted: 12/20/2023] [Indexed: 01/15/2024]
Abstract
The present study aims to establish the morphological, morphometric, and immunostaining patterns of the steroidogenic enzymes 17β-HSD and 5α-reductase and androgen receptors (AR) during the prenatal development of the male gonad and epididymis of Cavia porcellus. Fetuses at 22, 25, 30, 40, 45, 50, and 60 days of gestation (DG) were used. Specimens were dissected and subjected to macroscopic, histological, histomorphometric, and immunohistochemical analyses. Genital and scrotal protrusions were identified in 22 DG embryos. Gonocytes were identified at 25 DG and the formation of primary testicular cords was observed at 30 DG. Through anatomical evaluation, we observed differentiation of the epididymis into the head, body, and tail at 45 DG. During development, there is a progressive decrease in the diameters of the testicular cords and epididymal ducts. 17β-HSD enzyme immunostaining was observed in Leydig cells at all ages, while 5α-reductase was observed in Leydig cell cytoplasm and gonocytes at 40, 50, and 60 DG. AR shows gonocyte labeling at 30 DG. Thus, from the second trimester of pregnancy, it is possible to observe patterns of anatomical development, such as genital and scrotal prominence (22 DG), the appearance of gonocytes in the testicular cords at 25 DG, and the beginning of the organization of primary testicular cords at 30 DG, suggesting sexual differentiation. The 17β-HSD, 5α-reductase, and ARs play an essential role in sexual development and differentiation, presenting immunostaining at different reproductive process times.
Collapse
Affiliation(s)
- G A A Lemos
- School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - A C Santos
- School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - D C C Brito
- School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil; Laboratory of Manipulation of Oocytes and Ovarian Pre-Antral Follicles (LAMOFOPA), Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil
| | - M A S Novaes
- Laboratory of Manipulation of Oocytes and Ovarian Pre-Antral Follicles (LAMOFOPA), Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil
| | - A C Assis Neto
- School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil.
| |
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: 0.5] [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
|
Su J, Song Y, Yang Y, Li Z, Zhao F, Mao F, Wang D, Cao G. Study on the changes of LHR, FSHR and AR with the development of testis cells in Hu sheep. Anim Reprod Sci 2023; 256:107306. [PMID: 37541020 DOI: 10.1016/j.anireprosci.2023.107306] [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] [Received: 03/21/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 08/06/2023]
Abstract
The process of testis development in mammals is accompanied by the proliferation and maturation of Sertoli, Leydig and germ cells. Spermatogenesis depends on hormone regulation, which must bind to a receptor to exert its biological effects. The changes in Hu sheep testis cell composition and FSHR, LHR and AR expression during different developmental stages are unclear (newborn, puberty and adulthood). To address this, using single-cell RNA sequencing, we analyzed testis cell composition and hormone receptor expression changes during three important developmental stages of Hu sheep. We observed significant changes in the composition of somatic and germ cells in different Hu sheep testis developmental stages. Furthermore, we analyzed the FSHR, LHR and AR distribution and expression changes at three important periods and verified them by qRT-PCR and immunofluorescence. Our results suggest that after birth, the proportion of germ cells increased gradually, peaking in adulthood; the proportion of Sertoli cells decreased gradually, reaching the lowest in adulthood; and the proportion of Leydig cells increased and then decreased, reaching the lowest in adulthood. In addition, FSHR, LHR and AR are mainly located in Sertoli, Leydig and germ cells. LHR and FSHR expression decreased with increasing age, while AR expression increased and then decreased with increasing age.
Collapse
Affiliation(s)
- Jie Su
- Department of Medical Neurobiology, Inner Mongolia Medical University, Huhhot 010030, China; Inner Mongolia Key Laboratory of Basic Veterinary Science, Inner Mongolia Agriculture University, Hohhot 010018, China
| | - Yongli Song
- Research Center for Animal Genetic Resources of Mongolia Plateau, Inner Mongolia University, Huhhot 010021, China
| | - Yanyan Yang
- Inner Mongolia Academy of Agricultural & Animal Husbandry Sciences, Huhhot 010000, China
| | - Zhijun Li
- Department of Medical Neurobiology, Inner Mongolia Medical University, Huhhot 010030, China
| | - Feifei Zhao
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Inner Mongolia Agriculture University, Hohhot 010018, China
| | - Fei Mao
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Inner Mongolia Agriculture University, Hohhot 010018, China
| | - Daqing Wang
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Inner Mongolia Agriculture University, Hohhot 010018, China; Inner Mongolia Academy of Agricultural & Animal Husbandry Sciences, Huhhot 010000, China
| | - Guifang Cao
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Inner Mongolia Agriculture University, Hohhot 010018, China.
| |
Collapse
|
6
|
Overland MR, Li Y, Derpinghaus A, Aksel S, Cao M, Ladwig N, Cunha GR, Himelreich-Perić M, Baskin LS. Development of the human ovary: Fetal through pubertal ovarian morphology, folliculogenesis and expression of cellular differentiation markers. Differentiation 2023; 129:37-59. [PMID: 36347737 DOI: 10.1016/j.diff.2022.10.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/11/2022] [Accepted: 10/11/2022] [Indexed: 01/28/2023]
Abstract
A definition of normal human fetal and early postnatal ovarian development is critical to the ability to accurately diagnose the presence or absence of functional ovarian tissue in clinical specimens. Through assembling an extensive histologic and immunohistochemical developmental ontogeny of human ovarian specimens from 8 weeks of gestation through 16 years of postnatal, we present a comprehensive immunohistochemical mapping of normal protein expression patterns in the early fetal through post-pubertal human ovary and detail a specific expression-based definition of the early stages of follicular development. Normal fetal and postnatal ovarian tissue is defined by the presence of follicular structures and characteristic immunohistochemical staining patterns, including granulosa cells expressing Forkhead Box Protein L2 (FOXL2). However, the current standard array of immunohistochemical markers poorly defines ovarian stromal tissue, and additional work is needed to identify new markers to advance our ability to accurately identify ovarian stromal components in gonadal specimens from patients with disorders of sexual differentiation.
Collapse
Affiliation(s)
- Maya R Overland
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Yi Li
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Amber Derpinghaus
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Sena Aksel
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Mei Cao
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Nicholas Ladwig
- Department of Pathology, University of California, 505 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Gerald R Cunha
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA.
| | - Marta Himelreich-Perić
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000, Zagreb, Croatia
| | - Laurence S Baskin
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| |
Collapse
|
7
|
Cunha GR, Cao M, Aksel S, Derpinghaus A, Baskin LS. Mouse-human species differences in early testicular development and its implications. Differentiation 2023; 129:79-95. [PMID: 35667976 DOI: 10.1016/j.diff.2022.04.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/16/2022] [Accepted: 04/24/2022] [Indexed: 01/25/2023]
Abstract
The mouse has been used as a model of human organogenesis with the tacit assumption that morphogenetic and molecular mechanisms in mice are translatable to human organogenesis. While many morphogenetic and molecular mechanisms are shared in mice and humans, many anatomic, morphogenetic, and molecular differences have been noted. Two critical gaps in our knowledge prevent meaningful comparisons of mouse versus human testicular development: (a) human testicular development is profoundly under-represented in the literature, and (b) an absence of a detailed day-by-day ontogeny of mouse testicular development from E11.5 to E16.5 encompassing the ambisexual stage to seminiferous cord formation. To address these deficiencies, histologic and immunohistochemical studies were pursued in comparable stages of mouse and human testicular development with a particular emphasis on Leydig, Sertoli and myoid cells through review of the literature and new observations. For example, an androgen-receptor-positive testicular medulla is present in the developing human testis but not in the developing mouse testis. The human testicular medulla and associated mesonephros were historically described as the source of Sertoli cells in seminiferous cords. Consistent with this idea, the profoundly androgen receptor (AR)-positive human testicular medulla was shown to be a zone of mesenchymal to epithelial transition and a zone from which AR-positive cells appear to migrate into the human testicular cortex. While mouse Sertoli and Leydig cells have been proposed to arise from coelomic epithelium, Sertoli (SOX9) or Leydig (HSD3B1) cell markers are absent from the immediate coelomic zone of the developing human testis, perhaps because Leydig and Sertoli cell precursors are undifferentiated when they egress from the coelomic epithelium. The origin of mouse and human myoid cells remains unclear. This study provides a detailed comparison of the early stages of testicular development in human and mouse emphasizing differences in developmental processes.
Collapse
Affiliation(s)
- Gerald R Cunha
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA.
| | - Mei Cao
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Sena Aksel
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Amber Derpinghaus
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Laurence S Baskin
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| |
Collapse
|
8
|
Aksel S, Cao M, Derpinghaus A, Baskin LS, Cunha GR. Ontogeny of mouse Sertoli, Leydig and peritubular myoid cells from embryonic day 10 to adulthood. Differentiation 2023; 129:96-108. [PMID: 35317954 DOI: 10.1016/j.diff.2022.02.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 01/25/2023]
Abstract
We present a comprehensive description of the differentiating somatic cell types (Sertoli, Leydig, and peritubular myoid cells) of the mouse testis from embryonic day 10.5 (E10.5) to adulthood, postnatal day 60 (P60). Immunohistochemistry was used to analyze expression of: Sox9 (a Sertoli cell marker), 3βHSD-1 (a fetal Leydig cell marker), 3βHSD-6 (an adult Leydig cell marker), α-actin (a peritubular myoid cell marker), and androgen receptor (a marker of all three somatic cell types). The temporal-spatial expression of these markers was used to interrogate findings of earlier experimental studies on the origin of Sertoli, Leydig and peritubular myoid cells, as well as extend previous descriptive studies across a broader developmental period (E10.5-P60). Such comparisons demonstrate inconsistencies that require further examination and raise questions regarding conservation of developmental mechanisms across higher vertebrate species.
Collapse
Affiliation(s)
- Sena Aksel
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Mei Cao
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Amber Derpinghaus
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Laurence S Baskin
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Gerald R Cunha
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA.
| |
Collapse
|
9
|
Malolina EA, Galiakberova AA, Dashinimaev EB, Kulibin AY. ESTABLISHMENT OF A PURE CULTURE OF IMMATURE SERTOLI CELLS BY PDGFRA STAINING AND CELL SORTING. Mol Reprod Dev 2022; 89:243-255. [PMID: 35478364 DOI: 10.1002/mrd.23574] [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: 12/06/2021] [Revised: 04/02/2022] [Accepted: 04/25/2022] [Indexed: 11/10/2022]
Abstract
Sertoli cells are key somatic cells in the testis that form seminiferous tubules and support spermatogenesis. The isolation of pure Sertoli cells is important for their study. However, it is a difficult effort because of the close association of Sertoli cells with peritubular myoid cells surrounding seminiferous tubules. Here we propose a novel approach to the establishment of a pure Sertoli cell culture from immature mouse testes. It is based on the staining of testicular cells for platelet-derived growth factor receptor alpha (PDGFRA) followed by fluorescence-activated cell sorting and culturing of a PDGFRA-negative cell population. Cells positive for a Sertoli cell marker WT1 accounted for more than 96% of cells in cultures from 6 and 12 dpp mice. The numbers of peritubular myoid cells identified by ACTA2 staining did not exceed 4%. Cells in the cultures were also positive for Sertoli cell proteins SOX9 and DMRT1. Amh and Hsd17b3 expression decreased and Ar and Gata1 expression increased in 12 dpp cultures compared to 6 dpp cultures, which suggests that cultured Sertoli cells at least partially retained their differentiation status. This method can be employed in various applications including the analysis of differential gene expression and functional studies. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Ekaterina A Malolina
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334, Moscow, Russia
| | - Adelya A Galiakberova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, 117997, Russia
| | - Erdem B Dashinimaev
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334, Moscow, Russia.,Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, 117997, Russia
| | - Andrey Yu Kulibin
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334, Moscow, Russia
| |
Collapse
|
10
|
Cavariani MM, de Mello Santos T, Chuffa LGDA, Pinheiro PFF, Scarano WR, Domeniconi RF. Maternal Protein Restriction Alters the Expression of Proteins Related to the Structure and Functioning of the Rat Offspring Epididymis in an Age-Dependent Manner. Front Cell Dev Biol 2022; 10:816637. [PMID: 35517501 PMCID: PMC9061959 DOI: 10.3389/fcell.2022.816637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 04/05/2022] [Indexed: 11/16/2022] Open
Abstract
Nutrition is an environmental factor able to activate physiological interactions between fetus and mother. Maternal protein restriction is able to alter sperm parameters associated with epididymal functions. Since correct development and functioning of the epididymides are fundamental for mammalian reproductive success, this study investigated the effects of maternal protein restriction on epididymal morphology and morphometry in rat offspring as well as on the expression of Src, Cldn-1, AR, ER, aromatase p450, and 5α-reductase in different stages of postnatal epididymal development. For this purpose, pregnant females were allocated to normal-protein (NP—17% protein) and low-protein (LP—6% protein) groups that received specific diets during gestation and lactation. After weaning, male offspring was provided only normal-protein diet until the ages of 21, 44, and 120 days, when they were euthanized and their epididymides collected. Maternal protein restriction decreased genital organs weight as well as crown-rump length and anogenital distance at all ages. Although the low-protein diet did not change the integrity of the epididymal epithelium, we observed decreases in tubular diameter, epithelial height and luminal diameter of the epididymal duct in 21-day-old LP animals. The maternal low-protein diet changed AR, ERα, ERβ, Src 416, and Src 527 expression in offspring epididymides in an age-dependent manner. Finally, maternal protein restriction increased Cldn-1 expression throughout the epididymides at all analyzed ages. Although some of these changes did not remain until adulthood, the insufficient supply of proteins in early life altered the structure and functioning of the epididymis in important periods of postnatal development.
Collapse
|
11
|
Major AT, Estermann MA, Smith CA. Anatomy, Endocrine Regulation, and Embryonic Development of the Rete Testis. Endocrinology 2021; 162:6154516. [PMID: 33661305 DOI: 10.1210/endocr/bqab046] [Citation(s) in RCA: 13] [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: 12/06/2020] [Indexed: 12/23/2022]
Abstract
Reproduction in males requires the transfer of spermatozoa from testis tubules via the rete system to the efferent ductules, epididymis, and vas deferens. The rete therefore forms an essential bridging system between the testis and excurrent ducts. Yet the embryonic origin and molecular regulation of rete testis development is poorly understood. This review examines the anatomy, endocrine control, and development of the mammalian rete testis, focusing on recent findings on its molecular regulation, identifying gaps in our knowledge, and identifying areas for future research. The rete testis develops in close association with Sertoli cells of the seminiferous cords, although unique molecular markers are sparce. Most recently, modern molecular approaches such as global RNA-seq have revealed the transcriptional signature of rete cell precursors, pointing to at least a partial common origin with Sertoli cells. In the mouse, genes involved in Sertoli cell development or maintenance, such as Sox9, Wt1, Sf1, and Dmrt1, are also expressed in cells of the rete system. Rete progenitor cells also express unique markers, such as Pax8, E-cadherin, and keratin 8. These must directly or indirectly regulate the physical joining of testis tubules to the efferent duct system and confer other physiological functions of the rete. The application of technologies such as single-cell RNA-seq will clarify the origin and developmental trajectory of this essential component of the male reproductive tract.
Collapse
Affiliation(s)
- Andrew T Major
- Department of Anatomy and Developmental Biology, Monash Biomedical Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia
| | - Martin A Estermann
- Department of Anatomy and Developmental Biology, Monash Biomedical Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia
| | - Craig A Smith
- Department of Anatomy and Developmental Biology, Monash Biomedical Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia
| |
Collapse
|
12
|
Yaglova NV, Tsomartova DA, Obernikhin SS, Yaglov VV, Nazimova SV, Tsomartova ES, Chereshneva EV, Ivanova MY, Lomanovskaya TA. Differential Disrupting Effects of Prolonged Low-Dose Exposure to Dichlorodiphenyltrichloroethane on Androgen and Estrogen Production in Males. Int J Mol Sci 2021; 22:3155. [PMID: 33808818 PMCID: PMC8003643 DOI: 10.3390/ijms22063155] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/16/2021] [Accepted: 03/17/2021] [Indexed: 02/06/2023] Open
Abstract
Dichlorodiphenyltrichloroethane (DDT) is the most widespread, persistent pollutant and endocrine disruptor on the planet. Although DDT has been found to block androgen receptors, the effects of its low-dose exposure in different periods of ontogeny on the male reproductive system remain unclear. We evaluate sex steroid hormone production in the pubertal period and after maturation in male Wistar rats exposed to low doses of o,p'-DDT, either during prenatal and postnatal development or postnatal development alone. Prenatally and postnatally exposed rats exhibit lower testosterone production and increased estradiol and estriol serum levels after maturation, associated with the delayed growth of gonads. Postnatally exposed rats demonstrate accelerated growth of gonads and higher testosterone production in the pubertal period. In contrast to the previous group, they do not present raised estradiol production. All of the exposed animals exhibit a reduced conversion of progesterone to 17OH-progesterone after sexual maturation, which indicates putative attenuation of sex steroid production. Thus, the study reveals age-dependent outcomes of low-dose exposure to DDT. Prenatal onset of exposure results in the later onset of androgen production and the enhanced conversion of androgens to estrogens after puberty, while postnatal exposure induces the earlier onset of androgen secretion.
Collapse
Affiliation(s)
- Nataliya V. Yaglova
- Laboratory of Endocrine System Development, Federal State Budgetary Scientific Institution “Research Institute of Human Morphology”, 117418 Moscow, Russia; (D.A.T.); (S.S.O.); (V.V.Y.); (S.V.N.); (E.S.T.)
| | - Dibakhan A. Tsomartova
- Laboratory of Endocrine System Development, Federal State Budgetary Scientific Institution “Research Institute of Human Morphology”, 117418 Moscow, Russia; (D.A.T.); (S.S.O.); (V.V.Y.); (S.V.N.); (E.S.T.)
- Department of Histology, Cytology, and Embryology, Federal State Funded Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University, 119435 Moscow, Russia; (E.V.C.); (M.Y.I.); (T.A.L.)
| | - Sergey S. Obernikhin
- Laboratory of Endocrine System Development, Federal State Budgetary Scientific Institution “Research Institute of Human Morphology”, 117418 Moscow, Russia; (D.A.T.); (S.S.O.); (V.V.Y.); (S.V.N.); (E.S.T.)
| | - Valentin V. Yaglov
- Laboratory of Endocrine System Development, Federal State Budgetary Scientific Institution “Research Institute of Human Morphology”, 117418 Moscow, Russia; (D.A.T.); (S.S.O.); (V.V.Y.); (S.V.N.); (E.S.T.)
| | - Svetlana V. Nazimova
- Laboratory of Endocrine System Development, Federal State Budgetary Scientific Institution “Research Institute of Human Morphology”, 117418 Moscow, Russia; (D.A.T.); (S.S.O.); (V.V.Y.); (S.V.N.); (E.S.T.)
| | - Elina S. Tsomartova
- Laboratory of Endocrine System Development, Federal State Budgetary Scientific Institution “Research Institute of Human Morphology”, 117418 Moscow, Russia; (D.A.T.); (S.S.O.); (V.V.Y.); (S.V.N.); (E.S.T.)
- Department of Histology, Cytology, and Embryology, Federal State Funded Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University, 119435 Moscow, Russia; (E.V.C.); (M.Y.I.); (T.A.L.)
| | - Elizaveta V. Chereshneva
- Department of Histology, Cytology, and Embryology, Federal State Funded Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University, 119435 Moscow, Russia; (E.V.C.); (M.Y.I.); (T.A.L.)
| | - Marina Y. Ivanova
- Department of Histology, Cytology, and Embryology, Federal State Funded Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University, 119435 Moscow, Russia; (E.V.C.); (M.Y.I.); (T.A.L.)
| | - Tatiana A. Lomanovskaya
- Department of Histology, Cytology, and Embryology, Federal State Funded Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University, 119435 Moscow, Russia; (E.V.C.); (M.Y.I.); (T.A.L.)
| |
Collapse
|
13
|
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: 5.3] [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
|
14
|
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
|
15
|
Tian H, Huo Y, Zhang J, Ding S, Wang Z, Li H, Wang L, Lu M, Liu S, Qiu S, Zhang Q. Disruption of ubiquitin specific protease 26 gene causes male subfertility associated with spermatogenesis defects in mice†. Biol Reprod 2020; 100:1118-1128. [PMID: 30561524 DOI: 10.1093/biolre/ioy258] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 11/01/2017] [Accepted: 12/15/2018] [Indexed: 11/14/2022] Open
Abstract
Ubiquitin-specific protease 26 (USP26) is an X-linked gene exclusively expressed in the testis and codes for the USP26, a peptidase enzyme that belongs to the deubiquitinating enzyme family. Recent studies have indicated that mutations in USP26 affect spermatogenesis and are associated with male infertility in humans and mice. However, the exact role of USP26 in spermatogenesis and how it affects male reproduction remains unknown. In this study, we generated a conventional Usp26 knockout mouse model and found that deletion of Usp26 in male mice (Usp26-/Y) leads to significantly reduced pup numbers per litter and significantly increased intervals between two consecutive offspring. We also found that the serum follicle stimulating hormone and testosterone levels of adult Usp26-/Y mice were significantly decreased compared to those of Usp26+/Y mice. Histological examination results showed that Usp26-/Y mice had significantly increased percentage of abnormal seminiferous tubules at different ages. Flow cytometry results exhibited that Usp26-/Y mice had significantly reduced percentage of mature haploid cells in the testes compared to Usp26+/Y mice. Sperm counts in epididymis were also significantly declined in Usp26-/Y mice compared to those in Usp26+/Y mice. Immunohistochemistry and immunofluorescence staining and immunoprecipitation analysis results showed that USP26 and androgen receptor were co-localized in mouse testicular cells at different ages and they both had physiological interactions. All these results demonstrated that the loss of Usp26 affects spermatogenesis and hormone secretion and causes male subfertility. Our study also provides the evidence on the interactions between USP26 and androgen receptor in mouse testis, whereby pointing to a potential mechanism.
Collapse
Affiliation(s)
- Hong Tian
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China.,Research Center of Reproductive Medicine, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Yongwei Huo
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China.,Research Center of Reproductive Medicine, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Jie Zhang
- Dalian Municipal Women and Children's Medical Center, Dalian, Liaoning, China
| | - Shangshu Ding
- Research Center of Reproductive Medicine, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Zhiyong Wang
- Research Center of Reproductive Medicine, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China.,The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Hecheng Li
- Research Center of Reproductive Medicine, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China.,The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Lirong Wang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China.,Research Center of Reproductive Medicine, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Ming Lu
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Sen Liu
- Department of Structural & Cellular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Shudong Qiu
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China.,Research Center of Reproductive Medicine, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Qiuyang Zhang
- Department of Structural & Cellular Biology, Tulane University School of Medicine, New Orleans, LA, USA.,Tulane Center for Aging.,Tulane Cancer Center, Louisiana Cancer Research Consortium
| |
Collapse
|
16
|
Kamińska A, Marek S, Pardyak L, Brzoskwinia M, Pawlicki P, Bilińska B, Hejmej A. Disruption of androgen signaling during puberty affects Notch pathway in rat seminiferous epithelium. Reprod Biol Endocrinol 2020; 18:30. [PMID: 32299422 PMCID: PMC7161021 DOI: 10.1186/s12958-020-00582-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 03/04/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Onset of spermatogenesis at puberty is critically dependent on the activity of hypothalamic-pituitary-gonadal axis and testosterone production by Leydig cells. The aim of this study was to examine whether activation of Notch receptors and expression of Notch ligands and effector genes in rat seminiferous epithelium are controlled by androgen signaling during puberty. METHODS Peripubertal (5-week-old) Wistar rats received injections of flutamide (50 mg/kg bw) daily for 7 days to reduce androgen receptor (AR) signaling or a single injection of ethanedimethane sulphonate (EDS; 75 mg/kg bw) to reduce testosterone production. Gene and protein expressions were analyzed by real-time RT-PCR and western blotting, respectively, protein distribution by immunohistochemistry, and steroid hormone concentrations by enzyme-linked immunosorbent assay. Statistical analyses were performed using one-way ANOVA followed by Tukey's post hoc test or by Kruskal-Wallis test, followed by Dunn's test. RESULTS In both experimental models changes of a similar nature in the expression of Notch pathway components were found. Androgen deprivation caused the reduction of mRNA and protein expression of DLL4 ligand, activated forms of Notch1 and Notch2 receptors and HES1 and HEY1 effector genes (p < 0.05, p < 0.01, p < 0.001). In contrast, DLL1, JAG1 and HES5 expressions increased in seminiferous epithelium of both flutamide and EDS-treated rats (p < 0.05, p < 0.01, p < 0.001). CONCLUSIONS Androgens and androgen receptor signaling may be considered as factors regulating Notch pathway activity and the expression of Hes and Hey genes in rat seminiferous epithelium during pubertal development. Further studies should focus on functional significance of androgen-Notch signaling cross-talk in the initiation and maintenance of spermatogenesis.
Collapse
Affiliation(s)
- Alicja Kamińska
- Department of Endocrinology, Faculty of Biology, Institute of Zoology & Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387, Krakow, Poland
| | - Sylwia Marek
- Department of Endocrinology, Faculty of Biology, Institute of Zoology & Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387, Krakow, Poland
| | - Laura Pardyak
- Department of Endocrinology, Faculty of Biology, Institute of Zoology & Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387, Krakow, Poland
| | - Małgorzata Brzoskwinia
- Department of Endocrinology, Faculty of Biology, Institute of Zoology & Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387, Krakow, Poland
| | - Piotr Pawlicki
- Department of Endocrinology, Faculty of Biology, Institute of Zoology & Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387, Krakow, Poland
| | - Barbara Bilińska
- Department of Endocrinology, Faculty of Biology, Institute of Zoology & Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387, Krakow, Poland
| | - Anna Hejmej
- Department of Endocrinology, Faculty of Biology, Institute of Zoology & Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387, Krakow, Poland.
| |
Collapse
|
17
|
Kempinas WG, Borges CS, Leite GAA, Figueiredo TM, Gregory M, Cyr DG. Prenatal exposure to betamethasone causes intergenerational impairment of epididymal development in the rat. Andrology 2019; 7:719-729. [PMID: 31250541 DOI: 10.1111/andr.12657] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 04/30/2019] [Accepted: 05/05/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Studies on epididymal toxicology are scarce. Betamethasone (BM) is a glucocorticoid used in clinical practice for antenatal therapy. We previously reported changes to testicular morphology, altered sperm quality, and fertility in adult rats following intrauterine administration of BM. OBJECTIVES Given that high levels of corticosteroids during gestation lead to fetal androgen depletion, and the essential role of testosterone during epididymal development, here we investigated epididymal morphology and physiology in the F1 and F2 male offspring of female rats treated with BM during gestation. MATERIALS AND METHODS Pregnant rats were randomly divided into two experimental groups: control (saline vehicle, n = 11) and BM-treated group (0.1 mg/kg betamethasone 21-phosphate disodium, n = 13). Rats received an intramuscular injection of vehicle or BM on gestational days 12, 13, 18, and 19. This encompasses the beginning of the critical window of male rat reproductive tract development. A subset of three males from each litter (n = 5 litters/group) was used: One rat per litter was euthanized at puberty, one was euthanized at adulthood, while the others were mated with a non-treated female to obtain the F2 generation. The same protocol described for the F1 was applied for F2, except for the mating protocol. RESULTS In both F1 and F2 generations, prenatal BM exposure resulted in delayed differentiation of the cauda epididymal epithelium, characterized by increased cribriform appearance on PND 45, and displayed weaker or non-detectable Cx43 immunostaining. Furthermore, in the F1 generation only, immunostaining of TP63, a transcription factor expressed in basal cells, appeared more intense with a greater number of TP63-positive cells observed in the cauda epididymis. In adults, the epithelial area was reduced in the F1 BM rats. The contractile activity of isolated epididymal ducts was comparable between groups. DISCUSSION AND CONCLUSION Prenatal BM exposure leads to intergenerational impairment in the development and structure of the rat epididymis.
Collapse
Affiliation(s)
- W G Kempinas
- Laboratory of Reproductive and Developmental Biology and Toxicology, Morphology Department, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, Brazil
| | - C S Borges
- Laboratory of Reproductive and Developmental Biology and Toxicology, Morphology Department, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, Brazil
| | - G A A Leite
- Laboratory of Reproductive and Developmental Biology and Toxicology, Morphology Department, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, Brazil
| | - T M Figueiredo
- Laboratory of Reproductive and Developmental Biology and Toxicology, Morphology Department, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, Brazil
| | - M Gregory
- Laboratory for Reproductive Toxicology, INRS-Institut Armand-Frappier, University of Quebec, Laval, QC, Canada
| | - D G Cyr
- Laboratory for Reproductive Toxicology, INRS-Institut Armand-Frappier, University of Quebec, Laval, QC, Canada
| |
Collapse
|
18
|
da Cunha de Medeiros P, Samelo RR, Silva APG, da Silva Araujo Santiago M, Duarte FA, de Castro ÍB, Perobelli JE. Prepubertal exposure to low doses of sodium arsenite impairs spermatogenesis and epididymal histophysiology in rats. ENVIRONMENTAL TOXICOLOGY 2019; 34:83-91. [PMID: 30291770 DOI: 10.1002/tox.22660] [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] [Received: 07/06/2018] [Revised: 09/14/2018] [Accepted: 09/16/2018] [Indexed: 06/08/2023]
Abstract
For the first time, juvenile toxicity of inorganic arsenic (As) was investigated in male rats, focusing on reproductive effects. As is a metalloid naturally occurring in the environment, being the inorganic forms the most toxics. Contaminated drinking water and agricultural products are the main prospectors of intoxication for general population. In the present study, Wistar male rats (21 days old) were distributed into three groups (n = 10/group): control (received vehicle-filtered drinking water), As1 (received AsNaO2 at 0.01 mg L-1 ) and As2 (received AsNaO2 at 10 mg L-1 ). The animals were euthanized on PND 53. Testicular damages increased in As1 and As2 compared to control (ie, presence of vacuolization, acidophilic cells, and epithelium degeneration). Testicular interstitium of As1 and As2 presented fluid's increase and intense inflammatory infiltration. In the epididymis there was reduction of sperm amount in the lumen, besides epithelium areas presenting cribriform aspect in As1 and As2, exfoliation of cells in the light (in As1) and vacuoles (in As2). In epididymis interstitium, inflammatory infiltrates were observed in initial segment of As1 and As2. AsNaO2 changed immunolabeling pattern for androgen receptor in epididymis of As2, although serum testosterone levels was statistically comparable to control. Mass spectrometry revealed higher As concentrations in testis and epididymis of As2 compared to As1 and Control. These results indicate compromise of spermatogenesis and epididymal histophysiology in AsNaO2 -treated animals, possibly impairing sperm quality and fertility in long-term, even at low levels of exposure. Investigations about the reversibility of reproductive damages are necessary to better understand the mechanisms of As reproductive toxicity.
Collapse
Affiliation(s)
- Paloma da Cunha de Medeiros
- Laboratório de Toxicologia Experimental-LATOEX, Departamento de Ciências do Mar, Universidade Federal de São Paulo (UNIFESP), Campus Baixada Santista, Santos, São Paulo, Brazil
| | - Ricardo Rodrigues Samelo
- Laboratório de Toxicologia Experimental-LATOEX, Departamento de Ciências do Mar, Universidade Federal de São Paulo (UNIFESP), Campus Baixada Santista, Santos, São Paulo, Brazil
| | - Ana Priscila Gomes Silva
- Laboratório de Toxicologia Experimental-LATOEX, Departamento de Ciências do Mar, Universidade Federal de São Paulo (UNIFESP), Campus Baixada Santista, Santos, São Paulo, Brazil
| | - Marcella da Silva Araujo Santiago
- Laboratório de Toxicologia Experimental-LATOEX, Departamento de Ciências do Mar, Universidade Federal de São Paulo (UNIFESP), Campus Baixada Santista, Santos, São Paulo, Brazil
| | - Fabio Andrei Duarte
- Departamento de Química, Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Ítalo Braga de Castro
- Departamento de Ciências do Mar, Universidade Federal de São Paulo (UNIFESP), Campus Baixada Santista, Santos, São Paulo, Brazil
| | - Juliana Elaine Perobelli
- Laboratório de Toxicologia Experimental-LATOEX, Departamento de Ciências do Mar, Universidade Federal de São Paulo (UNIFESP), Campus Baixada Santista, Santos, São Paulo, Brazil
| |
Collapse
|
19
|
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: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 03/21/2019] [Indexed: 12/16/2022] Open
Abstract
Sertoli cells are somatic cells present in seminiferous tubules which have essential roles in regulating spermatogenesis. Considering that each Sertoli cell is able to support a limited number of germ cells, the final number of Sertoli cells reached during the proliferative period determines sperm production capacity. Only immature Sertoli cells, which have not established the blood-testis barrier, proliferate. A number of hormonal cues regulate Sertoli cell proliferation. Among them, FSH, the insulin family of growth factors, activin, and cytokines action must be highlighted. It has been demonstrated that cAMP/PKA, ERK1/2, PI3K/Akt, and mTORC1/p70SK6 pathways are the main signal transduction pathways involved in Sertoli cell proliferation. Additionally, c-Myc and hypoxia inducible factor are transcription factors which participate in the induction by FSH of various genes of relevance in cell cycle progression. Cessation of proliferation is a pre-requisite to Sertoli cell maturation accompanied by the establishment of the blood-testis barrier. With respect to this barrier, the participation of androgens, estrogens, thyroid hormones, retinoic acid and opioids has been reported. Additionally, two central enzymes that are involved in sensing cell energy status have been associated with the suppression of Sertoli cell proliferation, namely AMPK and Sirtuin 1 (SIRT1). Among the molecular mechanisms involved in the cessation of proliferation and in the maturation of Sertoli cells, it is worth mentioning the up-regulation of the cell cycle inhibitors p21Cip1, p27Kip, and p19INK4, and of the gap junction protein connexin 43. A decrease in Sertoli cell proliferation due to administration of certain therapeutic drugs and exposure to xenobiotic agents before puberty has been experimentally demonstrated. This review focuses on the hormones, locally produced factors, signal transduction pathways, and molecular mechanisms controlling Sertoli cell proliferation and maturation. The comprehension of how the final number of Sertoli cells in adulthood is established constitutes a pre-requisite to understand the underlying causes responsible for the progressive decrease in sperm production that has been observed during the last 50 years in humans.
Collapse
|
20
|
Kleshchev MA, Alekhina TA, Osadchuk LV. SPERM QUALITY IN RATS PREDISPOSED TO THE MANIFESTATION OF CATATONIC REACTIONS. Vavilovskii Zhurnal Genet Selektsii 2018. [DOI: 10.18699/vj18.375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Catatonia is a psychopathological syndrome displayed as a motor disorder. Catatonia is a sign of many menthal disorders, particularly schizophrenia and depression, with a wide disrtibution in the human population. The GC (“genetic” and “catatonia”) rat strain was obtained from the Wistar rat strain by a long selection (78 generations) for the catatonic type of reaction and is a model of schizophrenic and depressive disorders in humans. It is known that selection for behavior including catatonic reactions results in neuroendocrine, reproductive and morphological changes in animals. However, the influence of selection for a catatonic reaction on the spermatogenic function of testes had not been studied. The aim of this study was to conduct a comparative investigation of sperm quality in rats of the GC and the Wistar strain. The epididymal sperm parameters (sperm count, sperm motility, sperm morphology) were measured, and body, testes and epididymal weight were determined at puberty (50 day of life) and at adulthood (90 day of life). The litter size of the GC and Wistar rats was determined. It was found that adult GC rats had a lower sperm count, sperm motility, testis weight, epydidymal weight and litter size compared to adult Wistar rats. However, at puberty, GC rats had a higher sperm count than the Wistar strain. Interstrain differences in sperm morphology were not found. It has been assumed that the changes of spermatogenic parameters in response to selection for catatonia are caused by changing the ontogenic pattern of testosterone secretion. In conclusion, the hereditary predisposition to catatonic reaction is associated with impaired sperm parameters in adult rats that reduces their chance to reproduction. The GC rat strain can be a perspective model for investigation of the relationship between the hereditary predisposition to catatonia and spermatogenesis
Collapse
|
21
|
Lee KH. Aberrant Expression of Cx Isoforms in the Adult Caput Epididymis exposed to Estradiol Benzoate or Flutamide at the Weaning. Dev Reprod 2017; 21:379-389. [PMID: 29354784 PMCID: PMC5769132 DOI: 10.12717/dr.2017.21.4.379] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 12/14/2017] [Accepted: 12/22/2017] [Indexed: 11/17/2022]
Abstract
Connexin (Cx) involves in the regulation of various physiological functions of tissue by forming a channel, a gap junction which allows direct cell-cell communication, between adjacent cells. The effect of a single subcutaneous treatment of estradiol benzoate (EB) or flutamide (Flu) at the weaning age on the expression of Cx isoforms in the adult caput epididymis was evaluated in this research. Using quantitative real-time PCR analysis, a low-dose of EB [0.015 μg/kg body weight (BW)] caused significant decreases of Cx30.3, Cx32, Cx40, Cx43, and Cx45 mRNA levels and no change of Cx26, Cx31, Cx31.1, Cx37 transcript levels. The treatment of a high-dose EB (1.5 μg/kg BW) resulted in reduced expression of Cx30.3, Cx31, Cx43, and Cx45 but increased expression of Cx37 and Cx40. Expression of all Cx isoforms examined, except Cx31, was significantly increased by the treatment of a low-dose Flu (500 μg/kg BW). However, the treatment of a high-dose Flu (5 mg/kg BW) led significant expressional suppression of Cx30.3, Cx31, Cx31.1, Cx32, Cx40, Cx43, and Cx45 but an increase of Cx37 transcript level. With the comparison of previous findings, the expression of Cx isoforms in the adult epididymis after the exposure to EB or Flu is likely differentially regulated in regional-specific and/or exposed postnatal age-specific manner.
Collapse
Affiliation(s)
- Ki-Ho Lee
- Dept. of Biochemistry and Molecular Biology, College of Medicine, Eulji University, Daejeon 34824, Korea
| |
Collapse
|
22
|
Lee KH. Expressional Changes of Connexin Isoform Genes in the Rat Caput Epididymis Exposed to Flutamide or Estradiol Benzoate at the Early Postnatal Age. Dev Reprod 2017; 21:317-325. [PMID: 29082347 PMCID: PMC5651698 DOI: 10.12717/dr.2017.21.3.317] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 09/24/2017] [Accepted: 09/26/2017] [Indexed: 01/30/2023]
Abstract
Direct communication between neighboring cells through connexin
(Cx)-based gap junction is a crucial biolo–gical manner to
regulate functions of a tissue consisting of multi-cell types. The present
research evaluated expressional changes of Cx isoforms in the
caput epididymis of adult rat exposed to estradiol benzoate (EB) or flutamide
(Flu) at the early postnatal age. A single subcutaneous administration of EB at
a low-dose [0.015 µg /kg body weight (BW)] or a high-dose (1.5 µg/kg BW) or Flu
at a low-dose (500 µg/kg BW) or a high-dose (5 mg/kg BW) was performed to an
animal at 1 week of age. Quantitative real-time PCR analysis was employed to
determine expressional changes of Cx isoforms. The transcript
levels of Cxs30.3 and 37 were decreased by a low-dose EB
treatment, while decreases of Cxs31, 31.1, 32, 40, and 45
transcript levels were observed with a low-dose EB treatment. The treatment of a
high-dose EB resulted in expressional reduction of Cxs30.3, 31,
31.1, 37, 40, 43, and 45. The Flu treatment at a low dose caused increases of
Cxs26, 37, and 40 transcript levels but decreases of
Cxs31.1, 43, and 45 transcript levels. Increases of
Cxs30.3, 31, 37, and 40 mRNA amounts were induced by a
high-dose Flu treatment. However, exposure to a high-dose Flu produced
expressional decreases of Cxs31.1, 32, and 43 in the adult
caput epididymis. These observations suggest that exposure to EB or Flu at the
neonatal period could lead to aberrant expression of Cx
isoforms in the adult caput epididymis.
Collapse
Affiliation(s)
- Ki-Ho Lee
- Department of Biochemistry and Molecular Biology, College of Medicine, Eulji University, Daejeon 301-746, Korea
| |
Collapse
|
23
|
Leite GAA, Figueiredo TM, Sanabria M, Dias AFMG, Silva PVE, Martins Junior ADC, Barbosa Junior F, Kempinas WDG. Ascorbic acid supplementation partially prevents the delayed reproductive development in juvenile male rats exposed to rosuvastatin since prepuberty. Reprod Toxicol 2017; 73:328-338. [DOI: 10.1016/j.reprotox.2017.07.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 05/23/2017] [Accepted: 07/12/2017] [Indexed: 11/30/2022]
|
24
|
Lee KH. Effects of Exposure to Estradiol Benzoate or Flutamide at the Weaning Age on Expression of Connexins in the Caudal Epididymis of Adult Rat. Dev Reprod 2016; 20:349-357. [PMID: 28144639 PMCID: PMC5270609 DOI: 10.12717/dr.2016.20.4.349] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 11/22/2016] [Accepted: 12/17/2016] [Indexed: 11/28/2022]
Abstract
The present research was chiefly designed to determine the effect of the treatment of estrogenic
agonist, estradiol benzoate (EB), or antiandrogenic compound, flutamide (Flu), at the weaning age on
the expression of connexin (Cx) isoforms in the caudal epididymis of adult male
rat. Animals were subcutaneously administrated with a single shot of either EB at a low-dose (0.015
µg of EB/kg body weight (BW)) or a high-dose (1.5 µg of EB/kg BW) or Flu at a low-dose (500 µg of
EB/kg BW) or a high-dose (5 mg of EB/kg BW). Expressional changes of Cx isoforms in
the adult caudal epididymis were examined by quantitative real-time PCR analysis. The treatment of a
low-dose EB caused significant increases of Cx30.3, Cx31,
Cx32, and Cx43 transcript levels but reduction of
Cx31.1, Cx37, and Cx45 expression. Exposure
to a high-dose EB resulted in very close responses observed in a low-dose EB treatment, except no
significant expressional change of Cx37 and a significant induction of
Cx40. Expression of all Cx isoforms, except
Cx45, was significantly increased by a low-dose Flu treatment. Expressional
increases of all Cx isoforms were detected by a high-dose Flu treatment. The
current study demonstrates that a single exposure to estrogenic or antiandrogenic compound during
the early postnatal developmental period is sufficient to disrupt normal expression of
Cx isoforms in the adult caudal epididymis.
Collapse
Affiliation(s)
- Ki-Ho Lee
- Department of Biochemistry and Molecular Biology, College of Medicine, Eulji University, Daejeon 301-746, Korea
| |
Collapse
|
25
|
Ribeiro CM, Silva EJR, Hinton BT, Avellar MCW. β-defensins and the epididymis: contrasting influences of prenatal, postnatal, and adult scenarios. Asian J Androl 2016; 18:323-8. [PMID: 26763543 PMCID: PMC4770510 DOI: 10.4103/1008-682x.168791] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
β-defensins are components of host defense, with antimicrobial and pleiotropic immuno-modulatory properties. Research over the last 15 years has demonstrated abundant expression of a variety of β-defensins in the postnatal epididymis of different species. A gradient of region- and cell-specific expression of these proteins is observed in the epithelium of the postnatal epididymis. Their secretion into the luminal fluid and binding to spermatozoa as they travel along the epididymis has suggested their involvement in reproduction-specific tasks. Therefore, continuous attention has been given to various β-defensins for their role in sperm function and fertility. Although β-defensins are largely dependent on androgens, the underlying mechanisms regulating their expression and function in the epididymis are not well understood. Recent investigation has pointed out to a new and interesting scenario where β-defensins emerge with a different expression pattern in the Wolffian duct, the embryonic precursor of the epididymis, as opposed to the adult epididymis, thereby redefining the concept concerning the multifunctional roles of β-defensins in the developing epididymis. In this review, we summarize some current views of β-defensins in the epididymis highlighting our most recent data and speculations on their role in the developing epididymis during the prenatal-to-postnatal transition, bringing attention to the many unanswered questions in this research area that may contribute to a better understanding of epididymal biology and male fertility.
Collapse
Affiliation(s)
| | | | | | - Maria Christina W Avellar
- Section of Experimental Endocrinology, Department of Pharmacology, Universidade Federal de São Paulo - Escola Paulista de Medicina, São Paulo, Brazil
| |
Collapse
|
26
|
Cruceño AAM, Aguilera-Merlo CI, Chaves EM, Mohamed FH. Epididymis of Viscacha (Lagostomus maximus maximus): A Morphological Comparative Study in Relation to Sexual Maturity. Anat Histol Embryol 2016; 46:73-84. [PMID: 27457370 DOI: 10.1111/ahe.12240] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 05/29/2016] [Indexed: 01/23/2023]
Abstract
The morphological variations and the androgen receptor (AR) expression were studied in viscacha epididymis in relation to sexual maturity. The animals were divided into immature, pre-pubertal and adult, according to their corporal weight and testicular histology. The epididymides were studied by light microscopy, immunohistochemistry for AR and morphometric analysis. In pre-pubertal and adult animals, four well-differentiated segments (initial, caput, corpus and cauda) were observed, while in immature animals, three segments were identified (initial-caput segment, corpus and cauda). In each segment, the structural parameters and the relative cell distribution were different between the groups. The serum testosterone levels of pre-pubertal and adults showed a very significant increase related to sexual maturity. The AR expression in epithelial and fibromuscular stromal cells was different between the groups. In conclusion, the present work demonstrates that the morphological characteristics of the viscacha epididymis vary while sexual maturity is reached, the development of initial and caput is subsequent to corpus and cauda development and the androgens might play an important role during this process.
Collapse
Affiliation(s)
- A A M Cruceño
- Histología, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis, Argentina
| | - C I Aguilera-Merlo
- Histología, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis, Argentina
| | - E M Chaves
- Histología, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis, Argentina
| | - F H Mohamed
- Histología, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis, Argentina
| |
Collapse
|
27
|
Administration of testosterone inhibits initiation of seminal tubule growth and decreases Sertoli cell number in the earliest period of rat's postnatal development. Folia Histochem Cytobiol 2016; 47:S149-54. [PMID: 20067888 DOI: 10.2478/v10042-009-0094-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Sertoli cell (SC) number determines testes size and their capacity to produce spermatozoa. In the rat SC proliferate until 15th postnatal day (PND). Their proliferation is stimulated by FSH and inhibited by estradiol, but the role for androgens is uncertain. In this study we analyzed the effects of testosterone administration on testes growth and SC number in relation to timing of the treatment. Male rats were injected with 2.5 mg of testosterone propionate (TP) from birth until 5th PND and autopsied either on 6th PND [TP1-5(6)] or on 16th PND [TP1-5(16)] (transient administration). Other rats received TP from birth until 15th PND [TP1-15] or between 5th and 15th PND [TP5-15] continuously and were autopsied on day 16th. Control groups (C) received vehicle. In the Cs serum level of estradiol was 20-fold higher (p<0.001) and FSH was 1,7-fold higher (p<0.05) on 6th PND than on 16th PND, while testosterone did not change. After TP blood level of testosterone increased 2200-fold on 6th PND (p<0.05), and 8-fold on 16th PND. In turn, continuous TP administrations resulted on 16th PND in the increase in testosterone serum level by 2000-times of C without influence on FSH. While the treatment from birth either during initial 5 days or continuously until 15th day decreased testicular weight (p<0.001), tubule length (p<0.05) and SC number (p<0.001), the treatment initiated on 5th PND had no effects. TP reduced serum estradiol level on 6th PND by 13-fold (p<0.01), but doubled it on 16th PND. CONCLUSION Neonatal rats secrete estradiol and FSH in the amounts greatly extending those presented during further development. Testosterone inhibits testicular growth and SC number acting during first 5 neonatal days by decreasing FSH secretion, but is not effective during further development. Direct inhibitory influence of testosterone or trough its increased aromatisation to estradiol beyond neonatal period may be responsible for sustained inhibition of testes growth and SC number during infancy.
Collapse
|
28
|
Abstract
Androgens such as testosterone are steroid hormones essential for normal male reproductive development and function. Mutations of androgen receptors (AR) are often found in patients with disorders of male reproductive development, and milder mutations may be responsible for some cases of male infertility. Androgens exert their action through AR and its signalling in the testis is essential for spermatogenesis. AR is not expressed in the developing germ cell lineage so is thought to exert its effects through testicular Sertoli and peri-tubular myoid (PTM) cells. AR signalling in spermatogenesis has been investigated in rodent models where testosterone levels are chemically supressed or models with transgenic disruption of AR. These models have pinpointed the steps of spermatogenesis that require AR signalling, specifically maintenance of spermatogonial numbers, blood-testis barrier integrity, completion of meiosis, adhesion of spermatids and spermiation, together these studies detail the essential nature of androgens in the promotion of male fertility.
Collapse
Affiliation(s)
- Laura O'Hara
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
| | - Lee B Smith
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
| |
Collapse
|
29
|
Lee KH. Exogenous exposure to estradiol benzoate or flutamide at the weaning age alters expression of connexin isoforms in the initial segment of male rat. Dev Reprod 2015; 19:43-51. [PMID: 25949209 DOI: 10.12717/devrep.2015.19.1.043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 02/23/2015] [Accepted: 03/10/2015] [Indexed: 11/17/2022]
Abstract
Connexin (Cx) is a complex which allows direct communication between neighboring cells via exchange of signaling molecules and eventually leads to functional harmony of cells in a tissue. The initial segment (IS) is an excurrent duct of male reproductive tract and expression of numerous genes in the IS are controlled by andevrepogens and estrogens. The effects of these steroid hormones on gene expression in the IS during postnatal development have not extensively examined. The present research investigated expressional modulation of Cx isoforms in the IS by exogenous exposure to estrogen agonist, estradiol benzoate (EB), or andevrepogen antagonist, flutamide (Flu), at weaning age. Two different doses of EB or Flu were subcutaneously administrated in 21-day old of male rats, and expressional changes of Cx isoforms in the adult IS were analyzed by quantitative real-time PCR. Treatment of a low-dose EB (0.015 μg/kg body weight) resulted in an increased expression of Cx31 gene and a decreased expression of Cx37 gene. A high-dose EB (1.5 μg/kg body weight) treatment caused an increase of Cx31 gene expression. Increased levels of Cx30.3 and Cx40 transcripts were observed with a low-dose Flu (500 μg/kg body weight) treatment. Treatment of high-dose Flu (50 mg/kg body weight) led to expressional increases of Cx30.3, 40, and 43 genes. Our previous and present findings suggest differential responsiveness on gene expression of Cx isoforms in the IS by andevrepogens and estrogens at different postnatal ages.
Collapse
Affiliation(s)
- Ki-Ho Lee
- Dept. of Biochemistry and Molecular Biology, College of Medicine, Eulji University, Daejeon 301-746, Korea
| |
Collapse
|
30
|
Lee KH. Exogenous exposure to estradiol benzoate or flutamide at the weaning age alters expression of connexin isoforms in the initial segment of male rat. Dev Reprod 2015. [PMID: 25949209 PMCID: PMC4415663 DOI: 10.12717/dr.2015.19.1.043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Connexin (Cx) is a complex which allows direct communication between neighboring cells via exchange of signaling molecules and eventually leads to functional harmony of cells in a tissue. The initial segment (IS) is an excurrent duct of male reproductive tract and expression of numerous genes in the IS are controlled by androgens and estrogens. The effects of these steroid hormones on gene expression in the IS during postnatal development have not extensively examined. The present research investigated expressional modulation of Cx isoforms in the IS by exogenous exposure to estrogen agonist, estradiol benzoate (EB), or androgen antagonist, flutamide (Flu), at weaning age. Two different doses of EB or Flu were subcutaneously administrated in 21-day old of male rats, and expressional changes of Cx isoforms in the adult IS were analyzed by quantitative real-time PCR. Treatment of a low-dose EB (0.015 μg/kg body weight) resulted in an increased expression of Cx31 gene and a decreased expression of Cx37 gene. A high-dose EB (1.5 μg/kg body weight) treatment caused an increase of Cx31 gene expression. Increased levels of Cx30.3 and Cx40 transcripts were observed with a low-dose Flu (500 μg/kg body weight) treatment. Treatment of high-dose Flu (50 mg/kg body weight) led to expressional increases of Cx30.3, 40, and 43 genes. Our previous and present findings suggest differential responsiveness on gene expression of Cx isoforms in the IS by androgens and estrogens at different postnatal ages.
Collapse
Affiliation(s)
- Ki-Ho Lee
- Dept. of Biochemistry and Molecular Biology, College of Medicine, Eulji University, Daejeon 301-746, Korea
| |
Collapse
|
31
|
Systematic study of stress-inducible protein 1 (Stip1) in male reproductive system and its expression during stress response. Gene 2015; 554:58-63. [DOI: 10.1016/j.gene.2014.10.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 09/22/2014] [Accepted: 10/10/2014] [Indexed: 11/20/2022]
|
32
|
Smith LB, Walker WH. The regulation of spermatogenesis by androgens. Semin Cell Dev Biol 2014; 30:2-13. [PMID: 24598768 DOI: 10.1016/j.semcdb.2014.02.012] [Citation(s) in RCA: 528] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Revised: 02/20/2014] [Accepted: 02/24/2014] [Indexed: 11/16/2022]
Abstract
Testosterone is essential for maintaining spermatogenesis and male fertility. However, the molecular mechanisms by which testosterone acts have not begun to be revealed until recently. With the advances obtained from the use of transgenic mice lacking or overexpressing the androgen receptor, the cell specific targets of testosterone action as well as the genes and signaling pathways that are regulated by testosterone are being identified. In this review, the critical steps of spermatogenesis that are regulated by testosterone are discussed as well as the intracellular signaling pathways by which testosterone acts. We also review the functional information that has been obtained from the knock out of the androgen receptor from specific cell types in the testis and the genes found to be regulated after altering testosterone levels or androgen receptor expression.
Collapse
Affiliation(s)
- Lee B Smith
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK.
| | - William H Walker
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, 204 Craft Avenue, Pittsburgh, PA 15261, USA.
| |
Collapse
|
33
|
Lucas TF, Nascimento AR, Pisolato R, Pimenta MT, Lazari MFM, Porto CS. Receptors and signaling pathways involved in proliferation and differentiation of Sertoli cells. SPERMATOGENESIS 2014; 4:e28138. [PMID: 25225624 DOI: 10.4161/spmg.28138] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 02/05/2014] [Accepted: 02/06/2014] [Indexed: 12/11/2022]
Abstract
The identification of the hormones and other factors regulating Sertoli cell survival, proliferation, and maturation in neonatal, peripubertal, and pubertal life remains one of the most critical questions in testicular biology. The regulation of Sertoli cell proliferation and differentiation is thought to be controlled by cell-cell junctions and a set of circulating and local hormones and growth factors. In this review, we will focus on receptors and intracellular signaling pathways activated by androgen, follicle-stimulating hormone, thyroid hormone, activin, retinoids, insulin, insulin-like growth factor, relaxin, and estrogen, with special emphasis on estrogen receptors. Estrogen receptors activate intracellular signaling pathways that converge on cell cycle and transcription factors and play a role in the regulation of Sertoli cell proliferation and differentiation.
Collapse
Affiliation(s)
- Thaís Fg Lucas
- Section of Experimental Endocrinology; Department of Pharmacology; Escola Paulista de Medicina; Universidade Federal de São Paulo; INFAR; Vila Clementino; São Paulo, SP Brazil
| | - Aline R Nascimento
- Section of Experimental Endocrinology; Department of Pharmacology; Escola Paulista de Medicina; Universidade Federal de São Paulo; INFAR; Vila Clementino; São Paulo, SP Brazil
| | - Raisa Pisolato
- Section of Experimental Endocrinology; Department of Pharmacology; Escola Paulista de Medicina; Universidade Federal de São Paulo; INFAR; Vila Clementino; São Paulo, SP Brazil
| | - Maristela T Pimenta
- Section of Experimental Endocrinology; Department of Pharmacology; Escola Paulista de Medicina; Universidade Federal de São Paulo; INFAR; Vila Clementino; São Paulo, SP Brazil
| | - Maria Fatima M Lazari
- Section of Experimental Endocrinology; Department of Pharmacology; Escola Paulista de Medicina; Universidade Federal de São Paulo; INFAR; Vila Clementino; São Paulo, SP Brazil
| | - Catarina S Porto
- Section of Experimental Endocrinology; Department of Pharmacology; Escola Paulista de Medicina; Universidade Federal de São Paulo; INFAR; Vila Clementino; São Paulo, SP Brazil
| |
Collapse
|
34
|
Delayed reproductive development in pubertal male rats exposed to the hypolipemiant agent rosuvastatin since prepuberty. Reprod Toxicol 2014; 44:93-103. [PMID: 24440231 DOI: 10.1016/j.reprotox.2014.01.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 12/25/2013] [Accepted: 01/06/2014] [Indexed: 01/24/2023]
Abstract
Dyslipidemias are frequently found in children due to obesity, bad eating habits and the lack of physical exercises. Rosuvastatin acts as an HMG-CoA reductase inhibitor, decreasing total cholesterol and triglycerides. This study aimed to investigate initial sexual development and morphological aspect of the testis and epididymis in juvenile rats exposed to rosuvastatin since pre-puberty. Three groups were formed with newly weaned rats: control, whose rats received saline solution 0.9%, rosuvastatin at doses of 3 or 10 mg/kg daily by gavage, since post-natal day 21 until puberty onset. In the rosuvastatin-treated groups, the results demonstrated a trend toward a decrease in testosterone concentration, but below the significance level, as well as delays in both the age of puberty onset and in epididymal development. There were also testicular alterations that might be related to delayed puberty and decrease of serum testosterone. In conclusion, rosuvastatin administration to juvenile rats not only delayed puberty onset and epididymal development, but also impaired testicular and epididymal morphology.
Collapse
|
35
|
Rojas-García PP, Recabarren MP, Sir-Petermann T, Rey R, Palma S, Carrasco A, Perez-Marin CC, Padmanabhan V, Recabarren SE. Altered testicular development as a consequence of increase number of sertoli cell in male lambs exposed prenatally to excess testosterone. Endocrine 2013; 43:705-13. [PMID: 23076741 DOI: 10.1007/s12020-012-9818-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 10/09/2012] [Indexed: 10/27/2022]
Abstract
The reprograming effects of prenatal testosterone (T) treatment on postnatal reproductive parameters have been studied extensively in females of several species but similar studies in males are limited. We recently found that prenatal T treatment increases Sertoli cell number and reduced spermatogenesis in adult rams. If such disruptions are manifested early in life and involve changes in testicular paracrine environment remain to be explored. This study addresses the impact of prenatal T excess on testicular parameters in infant males, including Sertoli cell number and expression of critical genes [FSH receptor (FSHR), androgen receptor (AR), transforming growth factor beta 1 (TGFB1), 3 (TGFB3), transforming growth factor beta type 1 receptor, (TGFBR1), and anti-Müllerian hormone (AMH)] modulating testicular function. At 4 week of age, male lambs born to dams treated with 30 mg of T propionate twice weekly from day 30 to 90, followed by 40 mg of T propionate from day 90 to 120 of pregnancy (T-males), had a higher number of Sertoli cells/testis (P = 0.035) than control males (C-males) born to dams treated with the vehicle. While no differences were observed in the expression of FSHR and TGFB3, testicular TGFBR1 expression was found to be lower in T-males (P = 0.03) compared to C-males. Expression level of AMH, TGFB1, and AR also tended to be lower in T-males. These findings provide evidence that impact of fetal exposure to T excess is evident early in postnatal life, mainly characterized by an increase in Sertoli cell number. This could explain the testicular dysfunction observed in adult rams.
Collapse
Affiliation(s)
- Pedro P Rojas-García
- Laboratory of Animal Physiology and Endocrinology, Faculty of Veterinary Sciences, University of Concepción, Av. Vicente Mendez 595, Chillán, Chile.
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Schneider S, Marxfeld H, Gröters S, Buesen R, van Ravenzwaay B. Vinclozolin--no transgenerational inheritance of anti-androgenic effects after maternal exposure during organogenesis via the intraperitoneal route. Reprod Toxicol 2013; 37:6-14. [PMID: 23313085 DOI: 10.1016/j.reprotox.2012.12.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Revised: 12/19/2012] [Accepted: 12/20/2012] [Indexed: 10/27/2022]
Abstract
The goal of this study was to examine the potential transgenerational inheritance of anti-androgenic effects induced by Vinclozolin administered intraperitoneally to pregnant Wistar rats (Crl:WI[Han]). Dams were dosed with Vinclozolin at 0, 4 or 100mg/kg bw/d on gestation days 6-15. Male offspring of F1-F3 generations were bred with untreated females to yield F2-F4 offspring. No evident anti-androgenic effects were observed at 4mg/kg bw/d, but a case of hypospadias as well as delayed sexual maturation in F1 male offspring was observed as a sign of anti-androgenicity at 100mg/kg bw/d. However, F1-F3 males developed normally to sexual maturity and were able to mate and to generate healthy progeny. Sperm count, morphology and motility were not affected in F1-F4 generation male offspring. In conclusion, transgenerational inheritance of Vinclozolin's anti-androgenic effects was not evident in outbred Wistar rats.
Collapse
Affiliation(s)
- Steffen Schneider
- Experimental Toxicology and Ecotoxicology, BASF SE, 67056 Ludwigshafen, Germany
| | | | | | | | | |
Collapse
|
37
|
Hazra R, Corcoran L, Robson M, McTavish KJ, Upton D, Handelsman DJ, Allan CM. Temporal role of Sertoli cell androgen receptor expression in spermatogenic development. Mol Endocrinol 2012; 27:12-24. [PMID: 23160479 DOI: 10.1210/me.2012-1219] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Sertoli cell (SC) androgen receptor (AR) activity is vital for spermatogenesis. We created a unique gain-of-function transgenic (Tg) mouse model to determine the temporal role of SCAR expression in testicular development. The SC-specific rat Abpa promoter directed human Tg AR [Tg SC-specific AR (TgSCAR)] expression, providing strong premature postnatal AR immunolocalized to SC nuclei. Independent Tg lines revealed that TgSCAR dose dependently reduced postnatal and mature testis size (to 60% normal), whereas androgen-dependent mature seminal vesicle weights and serum testosterone levels remained normal. Total SC numbers were reduced in developing and mature TgSCAR testes, despite normal or higher Fshr mRNA and circulating FSH levels. Postnatal TgSCAR testes exhibited elevated levels of AR-regulated Rhox5 and Spinlw1 transcripts, and precocious SC function was demonstrated by early seminiferous tubular lumen formation and up-regulated expression of crucial SC tight-junction (Cldn11 and Tjp1) and phagocytic (Elmo1) transcripts. Early postnatal Amh expression was elevated but declined to normal levels in peripubertal-pubertal TgSCAR vs. control testes, indicating differential age-related regulation featuring AR-independent Amh down-regulation. TgSCAR induced premature postnatal spermatogenic development, shown by increased levels of meiotic (Dmc1 and Spo11) and postmeiotic (Capza3 and Prm1) germ cell transcripts, elevated meiotic-postmeiotic germ:Sertoli cell ratios, and accelerated spermatid development. Meiotic germ:Sertoli cell ratios were further increased in adult TgSCAR mice, indicating predominant SCAR-mediated control of meiotic development. However, postmeiotic germ:Sertoli cell ratios declined below normal. Our unique TgSCAR paradigm reveals that atypical SC-specific temporal AR expression provides a direct molecular mechanism for induction of precocious testicular development, leading to reduced adult testis size and decreased postmeiotic development.
Collapse
Affiliation(s)
- Rasmani Hazra
- ANZAC Research Institute, Concord Hospital, Sydney, New South Wales 2139, Australia
| | | | | | | | | | | | | |
Collapse
|
38
|
Alkafafy M, Sinowatz F. Prenatal development of the bovine epididymis: light microscopical, glycohistochemical and immunohistochemical studies. Acta Histochem 2012; 114:682-94. [PMID: 22204823 DOI: 10.1016/j.acthis.2011.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Revised: 12/05/2011] [Accepted: 12/07/2011] [Indexed: 12/12/2022]
Abstract
Prenatal development of the epididymis was studied in bovine fetuses ranging from 10 to 90cm crown-rump length (CRL) (75-285 pcd). The studies aimed to apply both glycohistochemistry and immunohistochemistry for the detection of the differentiation of the developing prenatal epididymis. Both conventional histological and histochemical techniques were applied on paraffin sections of the epididymis from different fetal stages. Establishment of the urogenital junction between the extra-testicular rete testis and the mesonephric duct, via the growing efferent ductules (ductuli efferentes) was first evident in fetuses with 10cm CRL. At the fetal age of 110 pcd (24cm CRL), the mesonephric duct began to lengthen and coil forming three distinct regions (caput, corpus and cauda). In addition to the macroscopical modifications in the extra-testicular excurrent duct system, histological differentiation involved both the tubular epithelial and the peritubular mesenchymal cells. The epithelium lining the efferent ductules was differentiated into ciliated and non-ciliated columnar cells. The simple epithelium of the epididymal duct increased in height and developed stereocilia on the apical surface. Additionally, some basal cells first appeared at 185 pcd (56cm CRL), within the epithelium lining the cauda only. Lectin histochemistry (WGA, PNA, GSA-I) showed early immunostaining in epithelium of the efferent ductules and in peritubular mesenchymal structures. Immunoreactivity for different proteins (S-100, fibroblast growth factor-1 and factor-2, angiotensin converting enzyme, laminin, alpha-smooth muscle actin) was evident, both in the epithelial and in the peritubular mesenchymal cells as early as at 75 pcd. On the basis of our histochemical observations, we conclude that both glycohistochemistry and immunohistochemistry are useful tools to demonstrate that the differentiation in the peritubular structures and efferent ductular epithelium begins earlier than other components.
Collapse
|
39
|
Tarulli GA, Stanton PG, Meachem SJ. Is the adult Sertoli cell terminally differentiated? Biol Reprod 2012; 87:13, 1-11. [PMID: 22492971 DOI: 10.1095/biolreprod.111.095091] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
New data have challenged the convention that the adult Sertoli cell population is fixed and unmodifiable. The Sertoli cell has two distinct functions: 1) formation of the seminiferous cords and 2) provision of nutritional and structural support to developing germ cells. For these to occur successfully, Sertoli cells must undergo many maturational changes between fetal and adult life, the main switches occurring around puberty, including the loss of proliferative activity and the formation of the blood-testis barrier. Follicle-stimulating hormone plays a key role in promoting Sertoli cell proliferation, while thyroid hormone inhibits proliferative activity in early postnatal life. Together these regulate the Sertoli-germ cell complement and sperm output in adulthood. By puberty, the Sertoli cell population is considered to be stable and unmodifiable by hormones. But there is mounting evidence that the size of the adult Sertoli cell population and its maturational status is modifiable by hormones and that Sertoli cells can gain proliferative ability in the spermatogenically disrupted hamster and human model. This new information demonstrates that the adult Sertoli cell population, at least in the settings of testicular regression in the hamster and impaired fertility in humans in vivo and from mice and men in vitro, is not a terminally differentiated population. Data from the hamster now show that the adult Sertoli cell population size is regulated by hormones. This creates exciting prospects for basic and clinical research in testis biology. The potential to replenish an adult Sertoli-germ cell complement to normal in a setting of infertility may now be realized.
Collapse
Affiliation(s)
- Gerard A Tarulli
- Prince Henry's Institute of Medical Research, Clayton, Victoria, Australia
| | | | | |
Collapse
|
40
|
Lydka M, Kopera-Sobota I, Kotula-Balak M, Chojnacka K, Zak D, Bilinska B. Morphological and functional alterations in adult boar epididymis: Effects of prenatal and postnatal administration of flutamide. Acta Vet Scand 2011; 53:12. [PMID: 21342526 PMCID: PMC3050768 DOI: 10.1186/1751-0147-53-12] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Accepted: 02/22/2011] [Indexed: 11/18/2022] Open
Abstract
Background The dynamic cross-talk between epididymal cells is hormonally regulated and, in part, through direct cell-to-cell interactions. To date, no information is available regarding possible impact of anti-androgens on the proteins involved in the gap junctional communication within the boar epididymis. Thus, a question arised whether prenatal or postnatal exposure to an anti-androgen flutamide alters the expression of gap junction protein - connexin43 (Cx43) and androgen receptor (AR) expression in the caput, corpus and cauda epididymis and leads to delayed effects on morphology and function of adult pig epididymis. Methods First two experimental groups received flutamide prenatally on gestational days 20-28 and 80-88 (GD20 and GD80) and further two groups were exposed to flutamide postanatally on days 2-10 and 90-98 after birth (PD2 and PD90). Epididymides were collected from adult boars. Routine histology was performed using hematoxylin-eosin staining. The expression of Cx43 and AR were analyzed using immunohistochemistry and Western blotting. Both analyses were supported by quantitative approaches to demonstrate the variations of the expression levels following the treatment. Apoptotic cells were identified using TUNEL assay. Results Histological examination revealed differences in epididymal morphology of flutamide-exposed boars when compared to controls. Scarce spermatic content were seen within the corpus and cauda lumina of GD20, PD2 and PD90 groups. Concomitantly, frequency of epididymal cell apoptosis was significantly higher (p < 0.05) after exposure to flutamide at GD20. Moreover, in GD20, PD2, and PD90 groups, significantly lower AR expression (p < 0.05) was found in the principal and basal cells of the corpus and cauda regions, while in the stromal cells AR expression was significantly reduced (p < 0.05) along the epididymal duct. Concomitantly, a decrease in Cx43 expression (p < 0.05) was noticed in the stromal cells of the cauda region of GD20 and PD2 groups. This indicates high sensitivity of the stromal cells to androgen withdrawal. Conclusions The region-specific alterations in the epididymis morphology and scarce spermatic content within the lumina of the corpus and cauda indicate that flutamide can induce delayed effects on the epididymal function of the adult boar by decrease in AR protein levels that results in altered androgen signaling. This may cause disturbances in androgen-dependent processes including Cx43 (de)regulation, however, we can not exclude the possibility that in response to flutamide decreased Cx43 expression may represent one mechanism responsible for functional disturbance of the boar epididymis.
Collapse
|
41
|
Rojas-García PP, Recabarren MP, Sarabia L, Schön J, Gabler C, Einspanier R, Maliqueo M, Sir-Petermann T, Rey R, Recabarren SE. Prenatal testosterone excess alters Sertoli and germ cell number and testicular FSH receptor expression in rams. Am J Physiol Endocrinol Metab 2010; 299:E998-E1005. [PMID: 20858754 DOI: 10.1152/ajpendo.00032.2010] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Exposure to excess testosterone (T) during fetal life has a profound impact on the metabolic and reproductive functions in the female's postnatal life. However, less is known about the effects of excess testosterone in males. The aim of the present study was to evaluate the impact (consequences) of an excess of T during fetal development on mature male testis. The testicular evaluation was by histological analysis and by determination of mRNA expression of the FSH receptor (FSH-R), transforming growth factor-β type I receptor (TβR-I), and two members of the TGF-β superfamily, transforming growth factor-β3 (TGFβ3) and anti-Müllerian hormone (AMH) in males born to mothers receiving an excess of T during pregnancy. At 42 wk of age, postpubertal males born to mothers treated with 30 mg of T propionate twice weekly from day 30 to 90, followed by 40 mg of T propionate from day 90 to 120 of pregnancy (T males), showed higher concentrations of FSH in response to a GnRH analog, a higher number of Sertoli cells/seminiferous tubule cross-section, and a lower number of germ cells/tubules (P < 0.05) than control males (C males) born to mothers treated with the vehicle. The mRNA expression of FSH-R and of TβR-I was higher in T males compared with C males (P < 0.05). Moreover, in T males, AMH expression level correlated negatively with the expression level of TGFβ3. In C males, this latter correlation was not observed. These results suggest that prenatal exposure to an excess of T can negatively modify some histological and molecular characteristics of the mature testis.
Collapse
Affiliation(s)
- Pedro P Rojas-García
- Faculty of Veterinary Sciences, Univ. of Concepción, Casilla 537, Chillán, Chile.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Patrão MTCC, Silva EJR, Avellar MCW. Androgens and the male reproductive tract: an overview of classical roles and current perspectives. ACTA ACUST UNITED AC 2009; 53:934-45. [DOI: 10.1590/s0004-27302009000800006] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2009] [Accepted: 11/14/2009] [Indexed: 11/22/2022]
Abstract
Androgens are steroid hormones that play key roles in the development and maintenance of male phenotype and reproductive function. These hormones also affect the function of several non-reproductive organs, such as bone and skeletal muscle. Endogenous androgens exert most of their effects by genomic mechanisms, which involve hormone binding to the androgen receptor (AR), a ligand-activated transcription factor, resulting in the modulation of gene expression. AR-induced non-genomic mechanisms have also been reported. A large number of steroidal and non-steroidal AR-ligands have been developed for therapeutic use, including the treatment of male hypogonadism (AR agonists) and prostate diseases (AR antagonists), among other pathological conditions. Here, the AR gene and protein structure, mechanism of action and AR gene homologous regulation were reviewed. The AR expression pattern, its in vivo regulation and physiological relevance in the developing and adult testis and epididymis, which are sites of sperm production and maturation, respectively, were also presented.
Collapse
|
43
|
Wang RS, Yeh S, Tzeng CR, Chang C. Androgen receptor roles in spermatogenesis and fertility: lessons from testicular cell-specific androgen receptor knockout mice. Endocr Rev 2009; 30:119-32. [PMID: 19176467 PMCID: PMC2662628 DOI: 10.1210/er.2008-0025] [Citation(s) in RCA: 328] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Androgens are critical steroid hormones that determine the expression of the male phenotype, including the outward development of secondary sex characteristics as well as the initiation and maintenance of spermatogenesis. Their actions are mediated by the androgen receptor (AR), a member of the nuclear receptor superfamily. AR functions as a ligand-dependent transcription factor, regulating expression of an array of androgen-responsive genes. Androgen and the AR play important roles in male spermatogenesis and fertility. The recent generation and characterization of male total and conditional AR knockout mice from different laboratories demonstrated the necessity of AR signaling for both external and internal male phenotype development. As expected, the male total AR knockout mice exhibited female-typical external appearance (including a vagina with a blind end and a clitoris-like phallus), the testis was located abdominally, and germ cell development was severely disrupted, which was similar to a human complete androgen insensitivity syndrome or testicular feminization mouse. However, the process of spermatogenesis is highly dependent on autocrine and paracrine communication among testicular cell types, and the disruption of AR throughout an experimental animal cannot answer the question about how AR in each type of testicular cell can play roles in the process of spermatogenesis. In this review, we provide new insights by comparing the results of cell-specific AR knockout in germ cells, peritubular myoid cells, Leydig cells, and Sertoli cells mouse models that were generated by different laboratories to see the consequent defects in spermatogenesis due to AR loss in different testicular cell types in spermatogenesis. Briefly, this review summarizes these results as follows: 1) the impact of lacking AR in Sertoli cells mainly affects Sertoli cell functions to support and nurture germ cells, leading to spermatogenesis arrest at the diplotene primary spermatocyte stage prior to the accomplishment of first meiotic division; 2) the impact of lacking AR in Leydig cells mainly affects steroidogenic functions leading to arrest of spermatogenesis at the round spermatid stage; 3) the impact of lacking AR in the smooth muscle cells and peritubular myoid cells in mice results in similar fertility despite decreased sperm output as compared to wild-type controls; and 4) the deletion of AR gene in mouse germ cells does not affect spermatogenesis and male fertility. This review tries to clarify the useful information regarding how androgen/AR functions in individual cells of the testis. The future studies of detailed molecular mechanisms in these in vivo animals with cell-specific AR knockout could possibly lead to useful insights for improvements in the treatment of male infertility, hypogonadism, and testicular dysgenesis syndrome, and in attempts to create safe as well as effective male contraceptive methods.
Collapse
Affiliation(s)
- Ruey-Sheng Wang
- Department of Pathology and Urology, George Whipple Laboratory for Cancer Research, University of Rochester Medical Center, Rochester, New York 14642, USA
| | | | | | | |
Collapse
|
44
|
Kopera I, Tuz R, Hejmej A, Schwarz T, Koczanowski J, Bilińska B. Immunolocalization of androgen receptor in the boar epididymis: the effect of GnRH agonist deslorelin. Reprod Domest Anim 2008; 44:266-72. [PMID: 18694429 DOI: 10.1111/j.1439-0531.2007.01054.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Epididymides from nine crossbred male pigs [Polish Landrace x (Duroc x Pietrain)] (n = 3 per each group) were used in this study to show whether there are any differences between androgen receptor (AR) distribution along epididymal duct of a GnRH agonist deslorelin-treated boars when compared to the control tissues. The active agent was administered by way of a subcutaneous controlled-release implant containing 4.7 mg deslorelin at 91 or 147 days of age respectively. Boars from two experimental groups and the control group were slaughtered at 175 day of age. Immunolocalization was performed using a polyclonal rabbit antiserum against the AR. In control boars, strong staining for AR was detected in nuclei of the epithelial (principal and basal) and stromal cells, whereas in boars treated with deslorelin the staining was confined to the principal cell nuclei. In those treated for 84 days, AR-immunostaining was weak or the principal cells were negative for the AR. Irrespective of the time from deslorelin insertion all stromal cells were immunonegative. The results demonstrate for the first time the effect of deslorelin on the distribution of the AR in the three regions of the boar epididymis. It is likely that stromal cells are more sensitive than epithelial cells to the regulation of AR expression by androgen. The morphological and functional alterations along the epididymal duct and lack of spermatozoa within the lumen after deslorelin treatment indicate that a potent GnRH agonist is likely responsible for an impairment of the microenvironment created by epididymal cells for sperm maturation and their storage.
Collapse
Affiliation(s)
- I Kopera
- Department of Endocrinology, Institute of Zoology, Jagiellonian University, Krakow, Poland
| | | | | | | | | | | |
Collapse
|
45
|
Kopera I, Tuz R, Kotula-Balak M, Schwarz T, Koczanowski J, Bilinska B. Morphofunctional alterations in testicular cells of deslorelin-treated boars: an immunohistochemical study. ACTA ACUST UNITED AC 2008; 309:117-26. [PMID: 18213611 DOI: 10.1002/jez.437] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
In this study we thoroughly scrutinized testes morphology and investigated whether treatment of recipient boars with gonadotropin-releasing hormone (GnRH)-agonist deslorelin could alter the expression of 3beta-hydroxysteroid dehydrogenase (3beta-HSD), luteinizing hormone receptors (LHRs), and androgen receptors (ARs) in testicular cells. An implant containing 4.7 mg of the GnRH-agonist deslorelin was subcutaneously inserted into crossbred male pigs at 91 and 147 days of age. Testicular traits, morphology of the testes, the proteins' expression, and testosterone concentration in blood plasma were analyzed in all boars after slaughter at 175 days of age. Histological analysis revealed significant alterations in both the interstitial tissue and seminiferous tubules of experimental animals after 28 and 84 days of deslorelin treatment. The intensity of the AR immunostaining within the testis appeared as a function of the severity of testicular dysgenesis. Time-dependent action of deslorelin on the expression of LHR and 3beta-HSD in Leydig cells was also detected. Staining for LHR and 3beta-HSD was very weak or the Leydig cells were immunonegative. Concomitantly, a significant decrease in plasma testosterone level was found in both groups of deslorelin-treated boars when compared with the control group. This is the first report showing the cellular distribution of AR, LHR, and 3beta-HSD in testicular cells of deslorelin-treated boars. It is concluded that morphological and immunohistochemical studies are important for the evaluation of testicular histoarchitecture and steroidogenic function. Subsequently, the endocrine control of reproduction in the GnRH-agonist deslorelin-treated males will be better understood.
Collapse
Affiliation(s)
- Ilona Kopera
- Department of Endocrinology and Tissue Culture, Institute of Zoology, Jagiellonian University, Kraków, Poland
| | | | | | | | | | | |
Collapse
|
46
|
Sonne SB, Kristensen DM, Novotny GW, Olesen IA, Nielsen JE, Skakkebæk NE, Rajpert-De Meyts E, Leffers H. Testicular dysgenesis syndrome and the origin of carcinoma in situ testis. ACTA ACUST UNITED AC 2008; 31:275-87. [DOI: 10.1111/j.1365-2605.2007.00855.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
47
|
Ramesh R, Pearl CA, At-Taras E, Roser JF, Berger T. Ontogeny of androgen and estrogen receptor expression in porcine testis: Effect of reducing testicular estrogen synthesis. Anim Reprod Sci 2007; 102:286-99. [PMID: 17157457 DOI: 10.1016/j.anireprosci.2006.10.025] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2006] [Accepted: 10/31/2006] [Indexed: 11/17/2022]
Abstract
Reducing endogenous estrogen leads to increased proliferation of porcine Sertoli cells during the first 2 months of life. The resulting increase in porcine Sertoli cell numbers is maintained through puberty. The reduced estrogen appears to be the direct hormonal mediator because essentially no changes are observed in other hormones. However, the mechanism for this effect on Sertoli cell proliferation is unknown. The objective of these studies was to evaluate estrogen receptors alpha and beta (ESR1 and ESR2) in conjunction with androgen receptor (AR) on Sertoli cells and other testicular cell types, as an initial step toward understanding how reduced estrogen leads to increased Sertoli cell numbers. Testis sections from treated animals (aromatase inhibition to decrease endogenous estrogen beginning at 1 week of age) and from littermate controls treated with vehicle were subjected to immunocytochemical labeling for ESR1, ESR2, and AR. Three observers scored Sertoli cells, interstitial cells, peritubular myoid cells, and germ cells for intensity of labeling (0: absent; 1+: weak; 2+: moderate; or 3+: strong labeling). AR in Sertoli cells was readily detected at 1 week of age, was very faint in 2-month vehicle controls, and labeling appeared to increase in 3-month vehicle controls. AR in Sertoli cells, interstitial cells, and apparently germ cells was increased in treated animals at 2 months of age compared with the vehicle controls. This increase was confirmed in western blots. ESR1 and ESR 2 were clearly present in Sertoli cells from 1-week-old animals; ESR in Sertoli cells generally decreased with age with the decrease more apparent for ESR2. ESR1 in Sertoli cells and peritubular myoid cells exhibited some treatment-related effects but reduction of endogenous estrogen did not appear to affect ESR2 in the boar testis. The observed alterations in AR and ESR1 may mediate the increases in Sertoli cell proliferation following inhibition of endogenous estrogen production or may reflect the altered function of the Sertoli cells and peritubular myoid cells.
Collapse
Affiliation(s)
- Revathi Ramesh
- Department of Animal Science, University of California Davis, Davis, CA 95616, USA
| | | | | | | | | |
Collapse
|
48
|
Scott HM, Hutchison GR, Mahood IK, Hallmark N, Welsh M, De Gendt K, Verhoeven G, O'Shaughnessy P, Sharpe RM. Role of androgens in fetal testis development and dysgenesis. Endocrinology 2007; 148:2027-36. [PMID: 17289843 DOI: 10.1210/en.2006-1622] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
This study sought to establish whether reduced androgen levels/action in the fetal rat testis induced by di(n-butyl) phthalate (DBP) contributes to dysgenetic features, namely reduced Sertoli cell number, occurrence of multinucleated gonocytes (MNG), and Leydig cell aggregation. Pregnant rats were administered treatments or cotreatments designed to manipulate testosterone levels [DBP, testosterone propionate (TP)] or action [flutamide, 7,12-dimethyl-benz[a]anthracene (DMBA)]. The aforementioned end points were analyzed and related to intratesticular testosterone (ITT) levels and peripheral androgen action (anogenital distance). Dysgenetic features were also evaluated in mice with inactivation of the androgen receptor (testicular feminized or ARKO mice). Exposure to DBP alone, or combined with flutamide, DMBA, or TP, resulted in reduced Sertoli cell number and ITT levels, as did exposure to TP alone; coadministration of DBP + TP caused the most severe reduction in both parameters. A positive correlation between ITT levels and Sertoli cell number was found (r = 0.791; P = 0.019). Similarly, exposure to DBP alone, or as a cotreatment, significantly increased occurrence of MNG and Leydig cell aggregation, and these were negatively correlated with ITT levels. Exposure to flutamide or DMBA alone had no significant effect on these dysgenetic end points. These findings suggest that reduced ITT decreases fetal Sertoli cell numbers and might be involved in Leydig cell aggregation and MNG. However, of these three end points, only Sertoli cell number was affected significantly in ARKO/testicular feminized mice with absent androgen action. Therefore, induction of MNG and Leydig cell aggregation might result from DBP-induced effects other than suppression of ITT levels.
Collapse
Affiliation(s)
- Hayley M Scott
- Medical Research Council Human Reproductive Sciences Unit, Centre for Reproductive Biology, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Merlet J, Racine C, Moreau E, Moreno SG, Habert R. Male fetal germ cells are targets for androgens that physiologically inhibit their proliferation. Proc Natl Acad Sci U S A 2007; 104:3615-20. [PMID: 17360691 PMCID: PMC1805536 DOI: 10.1073/pnas.0611421104] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In adulthood, the action of androgens on seminiferous tubules is essential for full quantitatively normal spermatogenesis and fertility. In contrast, their role in the fetal testis, and particularly in fetal germ cell development, remains largely unknown. Using testicular feminized (Tfm) mice, we investigated the effects of a lack of functional androgen receptor (AR) on fetal germ cells, also named gonocytes. We demonstrated that endogenous androgens/AR physiologically control normal gonocyte proliferation. We observed an increase in the number of gonocytes at 17.5 days postconception resulting from an increase in proliferative activity in Tfm mice. In a reciprocal manner, gonocyte proliferation is decreased by the addition of DHT in fetal testis organotypic culture. Furthermore, the AR coregulator Hsp90alpha (mRNA and protein) specifically expressed in gonocytes was down-regulated in Tfm mice at 15.5 days postconception. To investigate whether these effects could result from direct action of androgens on gonocytes, we collected pure gonocyte preparations and detected AR transcripts therein. We used an original model harboring a reporter gene that specifically reflects AR activity by androgens and clearly demonstrated the presence of a functional AR protein in fetal germ cells. These data provide in vivo and in vitro evidence of a new control of endogenous androgens on gonocytes identified as direct target cells for androgens. Finally, our results focus on a new pathway in the fetal testis during the embryonic period, which is the most sensitive to antiandrogenic endocrine disruptors.
Collapse
Affiliation(s)
- Jorge Merlet
- Laboratory of Differentiation and Radiobiology of the Gonads, Unit of Gametogenesis and Genotoxicity, Unité Mixte de Recherche-S 566, Université Denis Diderot Paris 7 and Commissariat à l'Énergie Atomique, Institut de Radiobiologie Cellulaire et Moléculaire, and Institut National de la Santé et de la Recherche Médicale Unité 566, F-92265 Fontenay-aux-Roses, France
| | - Chrystèle Racine
- Laboratory of Differentiation and Radiobiology of the Gonads, Unit of Gametogenesis and Genotoxicity, Unité Mixte de Recherche-S 566, Université Denis Diderot Paris 7 and Commissariat à l'Énergie Atomique, Institut de Radiobiologie Cellulaire et Moléculaire, and Institut National de la Santé et de la Recherche Médicale Unité 566, F-92265 Fontenay-aux-Roses, France
- *To whom correspondence should be addressed. E-mail:
| | - Evelyne Moreau
- Laboratory of Differentiation and Radiobiology of the Gonads, Unit of Gametogenesis and Genotoxicity, Unité Mixte de Recherche-S 566, Université Denis Diderot Paris 7 and Commissariat à l'Énergie Atomique, Institut de Radiobiologie Cellulaire et Moléculaire, and Institut National de la Santé et de la Recherche Médicale Unité 566, F-92265 Fontenay-aux-Roses, France
| | - Stéphanie G. Moreno
- Laboratory of Differentiation and Radiobiology of the Gonads, Unit of Gametogenesis and Genotoxicity, Unité Mixte de Recherche-S 566, Université Denis Diderot Paris 7 and Commissariat à l'Énergie Atomique, Institut de Radiobiologie Cellulaire et Moléculaire, and Institut National de la Santé et de la Recherche Médicale Unité 566, F-92265 Fontenay-aux-Roses, France
| | - René Habert
- Laboratory of Differentiation and Radiobiology of the Gonads, Unit of Gametogenesis and Genotoxicity, Unité Mixte de Recherche-S 566, Université Denis Diderot Paris 7 and Commissariat à l'Énergie Atomique, Institut de Radiobiologie Cellulaire et Moléculaire, and Institut National de la Santé et de la Recherche Médicale Unité 566, F-92265 Fontenay-aux-Roses, France
| |
Collapse
|
50
|
Mu X, Liu K, Kleymenova E, Sar M, Young SS, Gaido KW. Gene expression profiling of androgen receptor antagonists in the rat fetal testis reveals few common gene targets. J Biochem Mol Toxicol 2006; 20:7-17. [PMID: 16498641 DOI: 10.1002/jbt.20110] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The androgen receptor (AR) is expressed in the fetal testis; however, the role of AR in fetal testicular development is poorly understood. Disrupted AR activity and subsequent gene expression alterations may disturb developmental programming of the fetal testis and result in testicular abnormalities later in life. The present study was performed to examine global gene expression patterns in rat fetal testis following in utero exposure to various AR antagonists. Pregnant Sprague-Dawley rats were treated with flutamide (50 mg/kg/day), linuron (50 mg/kg/day), vinclozolin (200 mg/kg/day), p,p'-DDE (100 mg/kg/day) or corn oil vehicle by gavage daily from gestation day (GD) 12-19. Testes were isolated on GD 19, and AR immunostaining, histology, and global changes in gene expression were determined. There were no alterations in the pattern or expression level of AR and no apparent histological changes in the fetal testes in any treatment group. Microarray analysis using Dunnett's test with multiple testing correction revealed no significant gene expression alterations following exposure to flutamide, linuron, vinclozolin, and p,p'-DDE. A less stringent analysis yielded some chemical specific effects on gene expression, and these effects were further evaluated by real-time RT-PCR. Vinclozolin treatment reduced the expression of several genes involved in cholesterol biosynthesis, though the testosterone levels were unchanged in the fetal testes in any treatment group. In flutamide, linuron, and p,p'-DDE treatment groups, the expression of hemoglobin Y, beta-like embryonic chain (Hbb-y) was reduced. Myomesin 2 (Myom2) expression was increased following linuron treatment. Given the lack of a common set of genes and the absence of overt histopathology, we conclude that the fetal testis is not a major target for AR activity at this stage of development although some cell-type specific gene expression changes cannot be ruled out.
Collapse
Affiliation(s)
- Xueyan Mu
- CIIT Centers for Health Research, Research Triangle Park, NC 27709, USA
| | | | | | | | | | | |
Collapse
|