1
|
Cannarella R, Crafa A, Curto R, Condorelli RA, La Vignera S, Calogero AE. Obesity and male fertility disorders. Mol Aspects Med 2024; 97:101273. [PMID: 38593513 DOI: 10.1016/j.mam.2024.101273] [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: 02/22/2024] [Accepted: 04/03/2024] [Indexed: 04/11/2024]
Abstract
Often associated with obesity, male infertility represents a widespread condition that challenges the wellbeing of the couple. In this article, we provide a comprehensive and critical analysis of studies exploring the association between obesity and male reproductive function, to evaluate the frequency of this association, and establish the effects of increased body weight on conventional and biofunctional sperm parameters and infertility. In an attempt to find possible molecular markers of infertility in obese male patients, the numerous mechanisms responsible for infertility in overweight/obese patients are reviewed in depth. These include obesity-related functional hypogonadism, insulin resistance, hyperinsulinemia, chronic inflammation, adipokines, irisin, gut hormones, gut microbiome, and sperm transcriptome. According to meta-analytic evidence, excessive body weight negatively influences male reproductive health. This can occurr through a broad array of molecular mechanisms. Some of these are not yet fully understood and need to be further elucidated in the future. A better understanding of the effects of metabolic disorders on spermatogenesis and sperm fertilizing capacity is very useful for identifying new diagnostic markers and designing therapeutic strategies for better clinical management of male infertility.
Collapse
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy; Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Andrea Crafa
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Roberto Curto
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Rosita A Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy.
| |
Collapse
|
2
|
Hu Y, Luo NJ, Gan L, Xue HY, Luo KY, Zhang JJ, Wang XZ. Heat stress upregulates arachidonic acid to trigger autophagy in sertoli cells via dysfunctional mitochondrial respiratory chain function. J Transl Med 2024; 22:501. [PMID: 38797842 PMCID: PMC11129461 DOI: 10.1186/s12967-024-05182-y] [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: 12/18/2023] [Accepted: 04/07/2024] [Indexed: 05/29/2024] Open
Abstract
As a key factor in determining testis size and sperm number, sertoli cells (SCs) play a crucial role in male infertility. Heat stress (HS) reduces SCs counts, negatively impacting nutrient transport and supply to germ cells, and leading to spermatogenesis failure in humans and animals. However, how HS affects the number of SCs remains unclear. We hypothesized that changes in SC metabolism contribute to the adverse effects of HS. In this study, we first observed an upregulation of arachidonic acid (AA), an unsaturated fatty acid after HS exposure by LC-MS/MS metabolome detection. By increasing ROS levels, expression of KEAP1 and NRF2 proteins as well as LC3 and LAMP2, 100 µM AA induced autophagy in SCs by activating oxidative stress (OS). We observed adverse effects of AA on mitochondria under HS with a decrease of mitochondrial number and an increase of mitochondrial membrane potential (MMP). We also found that AA alternated the oxygen transport and absorption function of mitochondria by increasing glycolysis flux and decreasing oxygen consumption rate as well as the expression of mitochondrial electron transport chain (ETC) proteins Complex I, II, V. However, pretreatment with 5 mM NAC (ROS inhibitor) and 2 µM Rotenone (mitochondrial ETC inhibitor) reversed the autophagy induced by AA. In summary, AA modulates autophagy in SCs during HS by disrupting mitochondrial ETC function, inferring that the release of AA is a switch-like response, and providing insight into the underlying mechanism of high temperatures causing male infertility.
Collapse
Affiliation(s)
- Yu Hu
- Department of Reproductive Medicine, Department of Obstetrics and Gynecology, Affiliated Hospital of Zunyi Medical University, Affiliated Hospital of Zunyi Medical University, 563000, Zunyi, China
- Chongqing Key Laboratory of Forage and Herbivore, College of Veterinary Medicine, Southwest University, 400715, Chongqing, Beibei, China
| | - Nan Jian Luo
- Department of Preclinical Medicine, Zunyi Medical University, 563000, Zunyi, China
| | - Lu Gan
- Chongqing Key Laboratory of Forage and Herbivore, College of Veterinary Medicine, Southwest University, 400715, Chongqing, Beibei, China
| | - Hong Yan Xue
- Chongqing Key Laboratory of Forage and Herbivore, College of Veterinary Medicine, Southwest University, 400715, Chongqing, Beibei, China
| | - Ke Yan Luo
- Department of Reproductive Medicine, Department of Obstetrics and Gynecology, Affiliated Hospital of Zunyi Medical University, Affiliated Hospital of Zunyi Medical University, 563000, Zunyi, China
| | - Jiao Jiao Zhang
- Chongqing Key Laboratory of Forage and Herbivore, College of Veterinary Medicine, Southwest University, 400715, Chongqing, Beibei, China.
| | - Xian Zhong Wang
- Chongqing Key Laboratory of Forage and Herbivore, College of Veterinary Medicine, Southwest University, 400715, Chongqing, Beibei, China.
| |
Collapse
|
3
|
Hu Y, Li Q, Qian Z, BeiXiao, Luo K, Luo N. Joint Analysis of Genome-wide DNA Methylation and Transcription Sequencing Identifies the Role of BAX Gene in Heat Stress-Induced-Sertoli Cells Apoptosis. Reprod Sci 2024; 31:1311-1322. [PMID: 38180610 DOI: 10.1007/s43032-023-01430-6] [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: 10/05/2023] [Accepted: 12/12/2023] [Indexed: 01/06/2024]
Abstract
The problem of male infertility is a global health crisis and poses a serious threat to the well-being of families. Under heat stress (HS), the reduction of Sertoli cells (SCs) inhibits energy transport and nutrient supply to germ cells, leading to spermatogenesis failure. DNA methylation of genes is a central epigenetic regulatory mechanism in mammalian reproduction. However, it remains unclear how DNA methylation regulates gene expression in heat-stressed SCs. In this study, we investigated whether the decrease in SC levels during HS could be related to epigenetic DNA modifications. The cells exposed to HS showed changes in differential methylation cytosines and regions (DMCs/DMRs) and differential expression genes (DEGs), but not in global DNA methylations. One of the most important biological processes affected by HS is cell apoptosis induced by the intrinsic apoptotic signaling pathway (GO: 2,001,244, P < 0.05) by enrichment in the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). The joint analysis showed that several gene expressions in RNA-seq and WGBS overlapped and the shortlisted genes BAX, HSPH1, HSF1B, and BAG were strongly correlated with stress response and apoptosis. Methylation-specific PCR (MSP) and flow cytometry (FCM) analyzes showed that reduced promoter methylation and enhanced gene expression of BAX with a consequence of apoptosis. The activity of BAX, as well as an increase in its expression, is likely to result in a reduction of SCs population which could further impair ATP supply and adversely affect membrane integrity. These findings provide novel insights into the molecular mechanisms through which stressors cause male reproductive dysfunction and a new molecular etiology of male infertility.
Collapse
Affiliation(s)
- Yu Hu
- Department of Reproductive Medicine, Department of Obstetrics and Gynecology, Affiliated Hospital of Zunyi Medical University, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
| | - QingHan Li
- Department of Reproductive Medicine, Department of Obstetrics and Gynecology, Affiliated Hospital of Zunyi Medical University, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
| | - ZhengLi Qian
- Department of Reproductive Medicine, Department of Obstetrics and Gynecology, Affiliated Hospital of Zunyi Medical University, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
| | - BeiXiao
- Department of Reproductive Medicine, Department of Obstetrics and Gynecology, Affiliated Hospital of Zunyi Medical University, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
| | - KeYan Luo
- Department of Reproductive Medicine, Department of Obstetrics and Gynecology, Affiliated Hospital of Zunyi Medical University, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China.
| | - NanJian Luo
- Department of Preclinical Medicine, Zunyi Medical University, Zunyi, 563000, China.
| |
Collapse
|
4
|
Zhang S, Wei Y, Gao X, Song Y, Huang Y, Jiang Q. Unveiling the Ovarian Cell Characteristics and Molecular Mechanism of Prolificacy in Goats via Single-Nucleus Transcriptomics Data Analysis. Curr Issues Mol Biol 2024; 46:2301-2319. [PMID: 38534763 DOI: 10.3390/cimb46030147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/29/2024] [Accepted: 03/06/2024] [Indexed: 03/28/2024] Open
Abstract
Increases in litter size, which are influenced by ovulation, are responsible for between 74% and 96% of the economic value of genetic progress, which influences selection. For the selection and breeding of highly prolific goats, genetic mechanisms underlying variations in litter size should be elucidated. Here, we used single-nucleus RNA sequencing to analyze 44,605 single nuclei from the ovaries of polytocous and monotocous goats during the follicular phase. Utilizing known reference marker genes, we identified 10 ovarian cell types characterized by distinct gene expression profiles, transcription factor networks, and reciprocal interaction signatures. An in-depth analysis of the granulosa cells revealed three subtypes exhibiting distinct gene expression patterns and dynamic regulatory mechanisms. Further investigation of cell-type-specific prolificacy-associated transcriptional changes elucidated that "downregulation of apoptosis", "increased anabolism", and "upstream responsiveness to hormonal stimulation" are associated with prolificacy. This study provides a comprehensive understanding of the cell-type-specific mechanisms and regulatory networks in the goat ovary, providing insights into the molecular mechanisms underlying goat prolificacy. These findings establish a vital foundation for furthering understanding of the molecular mechanisms governing folliculogenesis and for improving the litter size in goats via molecular design breeding.
Collapse
Affiliation(s)
- Sanbao Zhang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning 530004, China
| | - Yirong Wei
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning 530004, China
| | - Xiaotong Gao
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning 530004, China
| | - Ying Song
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning 530004, China
| | - Yanna Huang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning 530004, China
| | - Qinyang Jiang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning 530004, China
| |
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
|
Wang H, Liu Z, Larsen M, Hastings R, Gunewardena S, Kumar TR. Identification of follicle-stimulating hormone-responsive genes in Sertoli cells during early postnatal mouse testis development. Andrology 2023; 11:860-871. [PMID: 37208854 DOI: 10.1111/andr.13459] [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: 12/16/2022] [Revised: 05/11/2023] [Accepted: 05/12/2023] [Indexed: 05/21/2023]
Abstract
BACKGROUND In the mouse testis, Sertoli cells rapidly divide during a narrow window of time pre-pubertally and differentiate thereafter. The number of Sertoli cells determines the testis size and germ cell-carrying capacity. Follicle-stimulating hormone (FSH) binds its cognate FSH-receptors expressed on Sertoli cells and acts as a mitogen to regulate their proliferation. Fshb-/- mutant adult male mice have reduced Sertoli cell number and testis size and reduced sperm number and motility. However, FSH-responsive genes in early postnatal mouse Sertoli cells are unknown. OBJECTIVES To identify FSH-responsive genes in early postnatal mouse Sertoli cells. MATERIALS AND METHODS A fluorescence-activated cell sorting method was developed to rapidly purify Sertoli cells from control and Fshb-/- mice carrying a Sox9 GfpKI allele. These pure Sertoli cells were used for large-scale gene expression analyses. RESULTS We show that mouse Sertoli cells rarely divide beyond postnatal day 7. Our in vivo BrdU labeling studies indicate loss of FSH results in a 30% reduction in Sertoli cell proliferation in mice at 5 days of age. Flowsorted GFP+ Sertoli cells with maximal Fshr expression were 97%-98% pure and mostly devoid of Leydig and germ cells as assessed by Taqman qPCR quantification of gene expression and immunolabeling of the corresponding cell-specific markers. Large-scale gene expression analysis identified several differentially regulated genes in flow-sorted GFP+ Sertoli cells obtained from testis of control and Fshb-/- mice at 5 days of age. The top 25 networks identified by pathway analysis include those related to the cell cycle, cell survival and most importantly, carbohydrate and lipid metabolism and molecular transport. DISCUSSION Several of the FSH-responsive genes identified in this study could serve as useful markers for Sertoli cell proliferation in normal physiology, toxicant-induced Sertoli cell/testis injury, and other pathological conditions. CONCLUSION Our studies reveal that FSH-regulates macromolecular metabolism and molecular transport networks of genes in early postnatal Sertoli cells most likely in preparation for establishment of functional associations with germ cells to successfully coordinate spermatogenesis.
Collapse
Affiliation(s)
- Huizhen Wang
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Zhenghui Liu
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Mark Larsen
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Richard Hastings
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Sumedha Gunewardena
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - T Rajendra Kumar
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
7
|
Bashawat M, Braun B, Müller K, Hermann B. Molecular phenotyping of domestic cat ( Felis catus) testicular cells across postnatal development - A model for wild felids. THERIOGENOLOGY WILD 2023; 2:100031. [PMID: 37461433 PMCID: PMC10350788 DOI: 10.1016/j.therwi.2023.100031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Molecular characterisation of testicular cells is a pivotal step towards a profound understanding of spermatogenesis and developing assisted reproductive techniques (ARTs) based on germline preservation. To enable the identification of testicular somatic and spermatogenic cell types in felids, we investigated the expression of five molecular markers at the protein level in testes from domestic cats (Felis catus) at different developmental phases (prepubertal, pubertal I and II, postpubertal I and II) classified by single-cell ploidy analysis. Our findings indicate a prominent co-labelling for two spermatogonial markers, UCHL1 and FOXO1, throughout postnatal testis development. Smaller subsets of UCHL1 or FOXO1 single-positive spermatogonia were also evident, with the FOXO1 single-positive spermatogonia predominantly observed in prepubertal testes. As expected, DDX4+ germ cells increased in numbers beginning in puberty, reaching a maximum at adulthood (post-pubertal phase), corresponding to the sequential appearance of labelled spermatogonia, spermatocytes and spermatids. Furthermore, we identified SOX9+ Sertoli cells and CYP17A1+ Leydig cells in all of the developmental groups. Importantly, testes of African lion (Panthera leo), Sumatran tiger (Panthera tigris sumatrae), Chinese leopard (Panthera pardus japonesis) and Sudan cheetah (Acinonyx jubatus soemmeringii) exhibited conserved labelling for UCHL1, FOXO1, DDX4, SOX9 and CYP17A1. The present study provides fundamental information about the identity of spermatogenic and somatic testicular cell types across felid development that will be useful for developing ART approaches to support endangered felid conservation.
Collapse
Affiliation(s)
- M. Bashawat
- Department of Biology, Humboldt University of Berlin, Invalidenstr. 42, D-10115 Berlin, Germany
- Department of Reproduction Biology, Leibniz Institute for Zoo and Wildlife Research, Alfred-Kowalke-Str. 17, D-10315 Berlin, Germany
| | - B.C. Braun
- Department of Reproduction Biology, Leibniz Institute for Zoo and Wildlife Research, Alfred-Kowalke-Str. 17, D-10315 Berlin, Germany
| | - K. Müller
- Department of Reproduction Biology, Leibniz Institute for Zoo and Wildlife Research, Alfred-Kowalke-Str. 17, D-10315 Berlin, Germany
| | - B.P. Hermann
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX 78249, USA
| |
Collapse
|
8
|
Guerra MT, Erthal RP, Punhagui-Umbelino APF, Trinque CM, Torres de Bari MA, Nunes TDM, Costa WF, Cleto PH, Fernandes GSA. Reproductive toxicity of maternal exposure to di(2-ethylhexyl)phthalate and butyl paraben (alone or in association) on both male and female Wistar offspring. J Appl Toxicol 2023; 43:242-261. [PMID: 35962557 DOI: 10.1002/jat.4377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/18/2022] [Accepted: 08/05/2022] [Indexed: 01/17/2023]
Abstract
Parabens and phthalates are commonly found as contaminants in human fluids and are able to provoke reproductive toxicity, being considered endocrine disruptors. To evaluate the effects of phthalate and paraben, alone or in combination, on reproductive development of the offspring, female pregnant Wistar rats were allocated in six experimental groups: Three control groups (gavage [CG], subcutaneous [CS], and gavage + subcutaneous) received corn oil as vehicle, and the remaining groups were exposed to di(2-ethylhexyl)phthalate (DEHP) (500 mg/kg, gavage), butyl paraben (BP) (100 mg/kg, subcutaneously), or MIX (DEHP + BP), from Gestational Day 12 until Postnatal Day (PND) 21. The following parameters were assessed on the offspring: anogenital distance and weight at PND 1, nipple counting at PND 13, puberty onset, estrous cycle, weights of reproductive and detoxifying organs, histological evaluation of reproductive organs, and sperm evaluations (counts, morphology, and motility). Female pups from MIX group presented reduced body weight at PND 1, lower AGD, and decreased endometrium thickness. Male animals showed decreased body weight at PND 1 and lower number of Sertoli cells on DEHP and MIX groups, MIX group revealed increase of abnormal seminiferous tubules, DEHP animals presented delayed preputial separation and higher percentage of immotile sperms, and BP males presented diminished number of Leydig cells. In conclusion, the male offspring was more susceptible to DEHP toxicity; even when mixed to paraben, the main negative effects observed seem to be due to antiandrogenic phthalate action. On the other hand, DEHP seems to be necessary to improve the effects of BP on reducing estrogen-dependent and increasing androgen-dependent events.
Collapse
Affiliation(s)
| | - Rafaela Pires Erthal
- Department of Pathological Sciences, Biological Sciences Center, State University of Londrina-UEL, Londrina, Brazil
| | | | - Camila Moreira Trinque
- Department of General Biology, Biological Sciences Center, State University of Londrina-UEL, Londrina, Brazil
| | | | | | - Wagner Ferrari Costa
- Department of General Biology, Biological Sciences Center, State University of Londrina-UEL, Londrina, Brazil
| | - Pedro Horácio Cleto
- Department of General Biology, Biological Sciences Center, State University of Londrina-UEL, Londrina, Brazil
| | | |
Collapse
|
9
|
Bhattacharya I, Dey S, Banerjee A. Revisiting the gonadotropic regulation of mammalian spermatogenesis: evolving lessons during the past decade. Front Endocrinol (Lausanne) 2023; 14:1110572. [PMID: 37124741 PMCID: PMC10140312 DOI: 10.3389/fendo.2023.1110572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 03/23/2023] [Indexed: 05/02/2023] Open
Abstract
Spermatogenesis is a multi-step process of male germ cell (Gc) division and differentiation which occurs in the seminiferous tubules of the testes under the regulation of gonadotropins - Follicle Stimulating Hormone (FSH) and Luteinising hormone (LH). It is a highly coordinated event regulated by the surrounding somatic testicular cells such as the Sertoli cells (Sc), Leydig cells (Lc), and Peritubular myoid cells (PTc). FSH targets Sc and supports the expansion and differentiation of pre-meiotic Gc, whereas, LH operates via Lc to produce Testosterone (T), the testicular androgen. T acts on all somatic cells e.g.- Lc, PTc and Sc, and promotes the blood-testis barrier (BTB) formation, completion of Gc meiosis, and spermiation. Studies with hypophysectomised or chemically ablated animal models and hypogonadal (hpg) mice supplemented with gonadotropins to genetically manipulated mouse models have revealed the selective and synergistic role(s) of hormones in regulating male fertility. We here have briefly summarized the present concept of hormonal control of spermatogenesis in rodents and primates. We also have highlighted some of the key critical questions yet to be answered in the field of male reproductive health which might have potential implications for infertility and contraceptive research in the future.
Collapse
Affiliation(s)
- Indrashis Bhattacharya
- Department of Zoology, School of Biological Science, Central University of Kerala, Kasaragod, Kerala, India
- *Correspondence: Arnab Banerjee, ; Indrashis Bhattacharya,
| | - Souvik Dey
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Arnab Banerjee
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS) Pilani, Goa, India
- *Correspondence: Arnab Banerjee, ; Indrashis Bhattacharya,
| |
Collapse
|
10
|
Neonatal corticosterone administration increases p27-positive Sertoli cell number and decreases Sertoli cell number in the testes of mice at prepuberty. Sci Rep 2022; 12:19402. [PMID: 36371473 PMCID: PMC9653474 DOI: 10.1038/s41598-022-23695-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 11/03/2022] [Indexed: 11/14/2022] Open
Abstract
Cortisol and corticosterone (CORT) are steroid, antistress hormones and one of the glucocorticoids in humans and animals, respectively. This study evaluated the effects of CORT administration on the male reproductive system in early life stages. CORT was subcutaneously injected at 0.36 (low-), 3.6 (middle-), and 36 (high-dosed) mg/kg body weight from postnatal day (PND) 1 to 10 in ICR mice. We observed a dose-dependent increase in serum CORT levels on PND 10, and serum testosterone levels were significantly increased only in high-dosed-CORT mice. Triiodothyronine levels were significantly higher in the low-dosed mice but lower in the middle- and high-dosed mice. However, testicular weights did not change significantly among the mice. Sertoli cell numbers were significantly reduced in low- and middle-dosed mice, whereas p27-positive Sertoli cell numbers increased in low- and middle-dosed mice. On PND 16, significant increases in testicular and relative testicular weights were observed in all-dosed-CORT mice. On PND 70, a significant decrease in testicular weight, Sertoli cell number, and spermatozoa count was observed. These results revealed that increased serum CORT levels in early life stages could induce p27 expression in Sertoli cells and terminate Sertoli cell proliferation, leading to decreased Sertoli cell number in mouse testes.
Collapse
|
11
|
Zhang B, Yan Z, Gao Y, Li J, Wang Z, Wang P, Yang Q, Huang X, Gun S. Integrated analysis of miRNA and mRNA expression profiles in testes of Landrace and Hezuo boars. Front Vet Sci 2022; 9:942669. [PMID: 36330159 PMCID: PMC9622794 DOI: 10.3389/fvets.2022.942669] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 09/27/2022] [Indexed: 11/21/2022] Open
Abstract
Precocious puberty is closely related to testicular development and spermatogenesis, and there is increasing evidence that miRNAs are involved in regulation of testicular development and spermatogenesis. However, little is known about the regulation of microRNAs (miRNAs) during precocious maturation in Hezuo (HZ) boars. In this study, serum Testosterone (T), Estradiol (E2), Follicle-stimulating hormone (FSH) and Luteinizing hormone (LH) levels were detected in HZ and Landrace (LC) boars in the postnatal period at 30, 90, 120, 180, and 240 days, and the testes of HZ and LC boars at 30 and 120 days were used for histological observation. In addition, we performed small RNA-Seq to identify miRNA at sexual immaturity (30-days-old) and maturity (120-days-old) of HZ boar testis (using LC boar as control) to reveal the key miRNA in regulation of precocious puberty. Hormone assay results showed that high levels of T, E2, FSH, and LH may be related to precocious sexual maturity of HZ boars, and that FSH may play an important function before sexual maturity. Histological observation showed that HZ boars developed earlier than LC boars and had reached sexual maturity at 120 days. Small RNA-Seq yielded a total of 359 exist miRNAs, 767 known miRNAs and 322 novel miRNAs in 12 samples; 549, 468, 133, and 247 differentially expressed (DE) miRNAs were identified between Ha vs. Hb, La vs. Lb, Ha vs. La, and Hb vs. Lb (log2 fold change >1 and p < 0.05). Enrichment analysis showed that target genes of these DE miRNAs were enriched in many gene ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) signaling pathways (such as PI3K-Akt, Hippo and Rap1 signaling pathways) were related to testicular development and spermatogenesis. Further screening, some miRNAs (such as ssc-miR-29b, ssc-miR-199b, ssc-miR-383, ssc-miR-149, ssc-miR-615, and ssc-miR-370) were possibly associated with precocious puberty. These results provide new light on miRNA regulatory mechanisms involved in precocious puberty.
Collapse
Affiliation(s)
- Bo Zhang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Zunqiang Yan
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Yi Gao
- Jilin Rongtai Agricultural Development Co., Ltd., Changchun, China
| | - Jiyou Li
- Gansu General Station of Animal Husbandry Technology Extension, Lanzhou, China
| | - Zike Wang
- Gansu General Station of Animal Husbandry Technology Extension, Lanzhou, China
| | - Pengfei Wang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Qiaoli Yang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Xiaoyu Huang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Shuangbao Gun
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
- Gansu Research Center for Swine Production Engineering and Technology, Lanzhou, China
- *Correspondence: Shuangbao Gun
| |
Collapse
|
12
|
Sepponen K, Lundin K, Yohannes DA, Vuoristo S, Balboa D, Poutanen M, Ohlsson C, Hustad S, Bifulco E, Paloviita P, Otonkoski T, Ritvos O, Sainio K, Tapanainen JS, Tuuri T. Steroidogenic factor 1 (NR5A1) induces multiple transcriptional changes during differentiation of human gonadal-like cells. Differentiation 2022; 128:83-100. [DOI: 10.1016/j.diff.2022.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 08/14/2022] [Accepted: 08/14/2022] [Indexed: 11/03/2022]
|
13
|
Fan Y, Xu Q, Qian H, Tao C, Wan T, Li Z, Yan W, Niu R, Huang Y, Chen M, Xu Q, Martin EM, Wang X, Qin Y, Lu C. High-fat diet aggravates prenatal low-dose DEHP exposure induced spermatogenesis disorder: Characterization of testicular metabolic patterns in mouse offspring. CHEMOSPHERE 2022; 298:134296. [PMID: 35301995 PMCID: PMC9533191 DOI: 10.1016/j.chemosphere.2022.134296] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/03/2022] [Accepted: 03/09/2022] [Indexed: 06/02/2023]
Abstract
Di-(2-ethylhexyl) phthalate (DEHP) is a widely used plasticizer and has been identified as a male prenatal reproductive toxicant. A high fat diet (HFD) has also been suggested as another potential disruptor of male reproductive function. Despite this potential synergism between DEHP exposure and HFD, little is known about the concomitant effects of prenatal DEHP and a subsequent HFD exposure on male offspring reproductive injury. Here we established a mouse model of prenatal exposure to DEHP (0.2 mg/kg/day) to assess the testicular development and spermatogenesis in offspring subjected to obesogenic diet during the pubertal period. Gross phenotype, hormone profiles and the testicular metabolome were analyzed to determine the underlying mechanism. We found that prenatal exposure to low-dose DEHP resulted in decreased sperm density, decreased testosterone (T) levels, increased luteinizing hormone (LH) levels and testicular germ cell apoptosis. Furthermore, these injury phenotypes were aggravated by pubertal HFD treatment. Testicular riboflavin and biotin metabolites were enriched implying their roles in contributing HFD to exacerbate offspring spermatogenesis disorders due to prenatal low-dose DEHP exposure. Our findings suggest that pubertal HFD exacerbates reproductive dysfunction associated with prenatal exposure to low-dose DEHP in male adult offspring.
Collapse
Affiliation(s)
- Yun Fan
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Department of Microbes and Infection, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Qiaoqiao Xu
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Hong Qian
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Chengzhe Tao
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Tingya Wan
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Zhi Li
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Wenkai Yan
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Rui Niu
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Yuna Huang
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Minjian Chen
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Qiujin Xu
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Chinese Research Academy of Environmental Sciences, Beijing, 100012, China
| | - Elizabeth M Martin
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, 111 TW Alexander Drive, NC, 27707, USA
| | - Xinru Wang
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Yufeng Qin
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Department of Microbes and Infection, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Chuncheng Lu
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
14
|
Molecular characterization and expression patterns of nuclear androgen receptors in the ovoviviparous black rockfish Sebastes schlegelii. AQUACULTURE AND FISHERIES 2022. [DOI: 10.1016/j.aaf.2022.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
15
|
Adamczewska D, Słowikowska-Hilczer J, Walczak-Jędrzejowska R. The Fate of Leydig Cells in Men with Spermatogenic Failure. Life (Basel) 2022; 12:570. [PMID: 35455061 PMCID: PMC9028943 DOI: 10.3390/life12040570] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/25/2022] [Accepted: 04/08/2022] [Indexed: 11/18/2022] Open
Abstract
The steroidogenic cells in the testicle, Leydig cells, located in the interstitial compartment, play a vital role in male reproductive tract development, maintenance of proper spermatogenesis, and overall male reproductive function. Therefore, their dysfunction can lead to all sorts of testicular pathologies. Spermatogenesis failure, manifested as azoospermia, is often associated with defective Leydig cell activity. Spermatogenic failure is the most severe form of male infertility, caused by disorders of the testicular parenchyma or testicular hormone imbalance. This review covers current progress in knowledge on Leydig cells origin, structure, and function, and focuses on recent advances in understanding how Leydig cells contribute to the impairment of spermatogenesis.
Collapse
Affiliation(s)
| | | | - Renata Walczak-Jędrzejowska
- Department of Andrology and Reproductive Endocrinology, Medical University of Lodz, 92-213 Lodz, Poland; (D.A.); (J.S.-H.)
| |
Collapse
|
16
|
Yokota S, Takeda K, Oshio S. Spatiotemporal Small Non-coding RNAs Expressed in the Germline as an Early Biomarker of Testicular Toxicity and Transgenerational Effects Caused by Prenatal Exposure to Nanosized Particles. FRONTIERS IN TOXICOLOGY 2022; 3:691070. [PMID: 35295114 PMCID: PMC8915876 DOI: 10.3389/ftox.2021.691070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 06/01/2021] [Indexed: 12/28/2022] Open
Abstract
In recent years, an apparent decline in human sperm quality has been observed worldwide. One in every 5.5 couples suffers from infertility, with male reproductive problems contributing to nearly 40% of all infertility cases. Although the reasons for the increasing number of infertility cases are largely unknown, both genetic and environmental factors can be contributing factors. In particular, exposure to chemical substances during mammalian male germ cell development has been linked to an increased risk of infertility in later life owing to defective sperm production, reproductive tract obstruction, inflammation, and sexual disorders. Prenatal exposure to nanomaterials (NMs) is no exception. In animal experiments, maternal exposure to NMs has been reported to affect the reproductive health of male offspring. Male germ cells require multiple epigenetic reprogramming events during their lifespan to acquire reproductive capacity. Given that spermatozoa deliver the paternal genome to oocytes upon fertilization, we hypothesized that maternal exposure to NMs negatively affects male germ cells by altering epigenetic regulation, which may in turn affect embryo development. Small non-coding RNAs (including microRNAs, PIWI-interacting RNAs, tRNA-derived small RNAs, and rRNA-derived small RNAs), which are differentially expressed in mammalian male germ cells in a spatiotemporal manner, could play important regulatory roles in spermatogenesis and embryogenesis. Thus, the evaluation of RNAs responsible for sperm fertility is of great interest in reproductive toxicology and medicine. However, whether the effect of maternal exposure to NMs on spermatogenesis in the offspring (intergenerational effects) really triggers multigenerational effects remains unclear, and infertility biomarkers for evaluating paternal inheritance have not been identified to date. In this review, existing lines of evidence on the effects of prenatal exposure to NMs on male reproduction are summarized. A working hypothesis of the transgenerational effects of sperm-derived epigenomic changes in the F1 generation is presented, in that such maternal exposure could affect early embryonic development followed by deficits in neurodevelopment and male reproduction in the F2 generation.
Collapse
Affiliation(s)
- Satoshi Yokota
- Division of Cellular and Molecular Toxicology, Center for Biological Safety and Research, National Institute of Health Sciences, Kawasaki, Japan
| | - Ken Takeda
- Division of Toxicology and Health Science, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Yamaguchi, Japan
| | - Shigeru Oshio
- Department of Hygiene Chemistry, School of Pharmaceutical Sciences, Ohu University, Koriyama, Japan
| |
Collapse
|
17
|
Wang JM, Li ZF, Yang WX. What Does Androgen Receptor Signaling Pathway in Sertoli Cells During Normal Spermatogenesis Tell Us? Front Endocrinol (Lausanne) 2022; 13:838858. [PMID: 35282467 PMCID: PMC8908322 DOI: 10.3389/fendo.2022.838858] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 02/01/2022] [Indexed: 01/18/2023] Open
Abstract
Androgen receptor signaling pathway is necessary to complete spermatogenesis in testes. Difference between androgen binding location in Sertoli cell classifies androgen receptor signaling pathway into classical signaling pathway and non-classical signaling pathway. As the only somatic cell type in seminiferous tubule, Sertoli cells are under androgen receptor signaling pathway regulation via androgen receptor located in cytoplasm and plasma membrane. Androgen receptor signaling pathway is able to regulate biological processes in Sertoli cells as well as germ cells surrounded between Sertoli cells. Our review will summarize the major discoveries of androgen receptor signaling pathway in Sertoli cells and the paracrine action on germ cells. Androgen receptor signaling pathway regulates Sertoli cell proliferation and maturation, as well as maintain the integrity of blood-testis barrier formed between Sertoli cells. Also, Spermatogonia stem cells achieve a balance between self-renewal and differentiation under androgen receptor signaling regulation. Meiotic and post-meiotic processes including Sertoli cell - Spermatid attachment and Spermatid development are guaranteed by androgen receptor signaling until the final sperm release. This review also includes one disease related to androgen receptor signaling dysfunction named as androgen insensitivity syndrome. As a step further ahead, this review may be conducive to develop therapies which can cure impaired androgen receptor signaling in Sertoli cells.
Collapse
|
18
|
Ben Maamar M, Nilsson EE, Skinner MK. Epigenetic transgenerational inheritance, gametogenesis and germline development†. Biol Reprod 2021; 105:570-592. [PMID: 33929020 PMCID: PMC8444706 DOI: 10.1093/biolre/ioab085] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/12/2021] [Accepted: 04/22/2021] [Indexed: 12/14/2022] Open
Abstract
One of the most important developing cell types in any biological system is the gamete (sperm and egg). The transmission of phenotypes and optimally adapted physiology to subsequent generations is in large part controlled by gametogenesis. In contrast to genetics, the environment actively regulates epigenetics to impact the physiology and phenotype of cellular and biological systems. The integration of epigenetics and genetics is critical for all developmental biology systems at the cellular and organism level. The current review is focused on the role of epigenetics during gametogenesis for both the spermatogenesis system in the male and oogenesis system in the female. The developmental stages from the initial primordial germ cell through gametogenesis to the mature sperm and egg are presented. How environmental factors can influence the epigenetics of gametogenesis to impact the epigenetic transgenerational inheritance of phenotypic and physiological change in subsequent generations is reviewed.
Collapse
Affiliation(s)
- Millissia Ben Maamar
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA, USA
| | - Eric E Nilsson
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA, USA
| | - Michael K Skinner
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA, USA
| |
Collapse
|
19
|
Abstract
Puberty is characterized by major changes in the anatomy and function of reproductive organs. Androgen activity is low before puberty, but during pubertal development, the testes resume the production of androgens. Major physiological changes occur in the testicular cell compartments in response to the increase in intratesticular testosterone concentrations and androgen receptor expression. Androgen activity also impacts on the internal and external genitalia. In target cells, androgens signal through a classical and a nonclassical pathway. This review addresses the most recent advances in the knowledge of the role of androgen signaling in postnatal male sexual development, with a special emphasis on human puberty.
Collapse
Affiliation(s)
- Rodolfo A Rey
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Histología, Embriología, Biología Celular y Genética, C1121ABG Buenos Aires, Argentina
| |
Collapse
|
20
|
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
|
21
|
Shah W, Khan R, Shah B, Khan A, Dil S, Liu W, Wen J, Jiang X. The Molecular Mechanism of Sex Hormones on Sertoli Cell Development and Proliferation. Front Endocrinol (Lausanne) 2021; 12:648141. [PMID: 34367061 PMCID: PMC8344352 DOI: 10.3389/fendo.2021.648141] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 05/17/2021] [Indexed: 12/30/2022] Open
Abstract
Sustaining and maintaining the intricate process of spermatogenesis is liable upon hormones and growth factors acting through endocrine and paracrine pathways. The Sertoli cells (SCs) are the major somatic cells present in the seminiferous tubules and are considered to be the main regulators of spermatogenesis. As each Sertoli cell supports a specific number of germ cells, thus, the final number of Sertoli cells determines the sperm production capacity. Similarly, sex hormones are also major regulators of spermatogenesis and they can determine the proliferation of Sertoli cells. In the present review, we have critically and comprehensively discussed the role of sex hormones and some other factors that are involved in Sertoli cell proliferation, differentiation and maturation. Furthermore, we have also presented a model of Sertoli cell development based upon the recent advancement in the field of reproduction. Hence, our review article provides a general overview regarding the sex hormonal pathways governing Sertoli cell proliferation and development.
Collapse
Affiliation(s)
| | - Ranjha Khan
- *Correspondence: Xiaohua Jiang, ; Ranjha Khan, ; Jie Wen,
| | | | | | | | | | - Jie Wen
- *Correspondence: Xiaohua Jiang, ; Ranjha Khan, ; Jie Wen,
| | - Xiaohua Jiang
- *Correspondence: Xiaohua Jiang, ; Ranjha Khan, ; Jie Wen,
| |
Collapse
|
22
|
Miyaso H, Nagahori K, Takano K, Omotehara T, Kawata S, Li ZL, Kuramasu M, Wu X, Ogawa Y, Itoh M. Neonatal maternal separation causes decreased numbers of sertoli cell, spermatogenic cells, and sperm in mice. Toxicol Mech Methods 2020; 31:116-125. [PMID: 33100103 DOI: 10.1080/15376516.2020.1841865] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neonatal maternal separation is an experimental model used to evaluate the effects of toxic stress in neonates, or early life stress. Although various physiological and psychological stresses during childhood have been reported, the effects of neonatal maternal separation on the male reproductive system remain unclear. Therefore, the present study evaluated the effects of neonatal maternal separation on the male reproductive system. In neonatal male ICR mice, maternal separation was performed for 0.5, 1, 2, and 4 hours/day, from postnatal day 1 to 10. At 10 weeks of age, the neonatal maternal separation mice exhibited decreases in both testicular weight and epididymal sperm number, along with various testicular morphological changes involving germ cells, Sertoli cells, and interstitial cells. Notably, neonatal maternal separation mice showed decreased numbers of Sertoli cells. Animals subjected to 0.5-, 1-, and 2-h/day neonatal maternal separation exhibited decreases in serum levels of testosterone but not in those of gonadotropin (luteinizing hormone and follicle-stimulating hormone). Together, these data showed that neonatal maternal separation in male mice causes decreased Sertoli cell numbers following puberty, resulting in subsequent decreased spermatogenic activity.
Collapse
Affiliation(s)
- Hidenobu Miyaso
- Department of Anatomy, Tokyo Medical University, Tokyo, Japan
| | - Kenta Nagahori
- Department of Anatomy, Tokyo Medical University, Tokyo, Japan
| | - Kaiya Takano
- Department of Anatomy, Tokyo Medical University, Tokyo, Japan
| | | | - Shinichi Kawata
- Department of Anatomy, Tokyo Medical University, Tokyo, Japan
| | - Zhong-Lian Li
- Department of Anatomy, Tokyo Medical University, Tokyo, Japan
| | - Miyuki Kuramasu
- Department of Anatomy, Tokyo Medical University, Tokyo, Japan
| | - Xi Wu
- Department of Anatomy, Tokyo Medical University, Tokyo, Japan
| | - Yuki Ogawa
- Department of Anatomy, Tokyo Medical University, Tokyo, Japan
| | - Masahiro Itoh
- Department of Anatomy, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
23
|
Shobana N, Kumar MK, Navin AK, Akbarsha MA, Aruldhas MM. Prenatal exposure to excess chromium attenuates transcription factors regulating expression of androgen and follicle stimulating hormone receptors in Sertoli cells of prepuberal rats. Chem Biol Interact 2020; 328:109188. [PMID: 32679048 DOI: 10.1016/j.cbi.2020.109188] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 06/06/2020] [Accepted: 06/25/2020] [Indexed: 01/06/2023]
Abstract
We have reported that gestational exposure to hexavalent chromium (CrVI) represses androgen receptor (Ar) and follicle stimulating hormone receptor (Fshr) in Sertoli cells (SCs) of adult rats, while the mechanism underlying remains obscure. We tested the hypothesis "transient gestational exposure to CrVI during the critical embryonic windows of testicular differentiation and growth may have adverse impact on transcription factors controlling the expression of Ar and Fshr in SCs of the F1 progeny". CrVI (K2Cr2O7) was given through drinking water (50 ppm, 100 ppm and 200 ppm), to pregnant rats from gestational day 9-14 (testicular differentiation) and 15 to 21 (prenatal differentiation and proliferation of SC); male progenies were sacrificed on postnatal day 30 (Completion of postnatal SC maturation). A significant increase in free radicals and decrease in enzymatic and non-enzymatic antioxidants were observed in SCs of experimental rats. Real time PCR and western blot data showed decreased expression of Ar, Fshr, Inhibin B, Transferrin, Androgen binding protein, Claudin 11 and Occludin in SCs of experimental rats; concentrations of lactate, pyruvate and retinoic acid also decreased. Serum FSH, luteinizing hormone and estradiol increased, whereas testosterone and prolactin decreased in experimental rats. Western blot detection revealed decreased levels of transcription factors regulating Fshr viz., USF-1, USF-2, SF-1, c-fos, c-jun and GATA 1, and those of Ar viz., Sp-1, ARA54, SRC-1 and CBP in experimental rats, whereas the levels of cyclinD1 and p53, repressors of Ar increased. ChIP assay detected decreased USF-1 and USF-2 binding to Fshr promoter, and binding of Sp-1 to Ar promoter. We conclude that gestational exposure to CrVI affects SC structure and function in F1 progeny by inducing oxidative stress and diminishing the expression of Ar and Fshr through attenuation of their specific transcriptional regulators and their interaction with the respective promoter.
Collapse
Affiliation(s)
- Navaneethabalakrishnan Shobana
- Department of Endocrinology, Dr.A.L.M Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, 600113, India
| | - Mani Kathiresh Kumar
- Department of Endocrinology, Dr.A.L.M Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, 600113, India
| | - Ajit Kumar Navin
- Department of Endocrinology, Dr.A.L.M Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, 600113, India
| | | | - Mariajoseph Michael Aruldhas
- Department of Endocrinology, Dr.A.L.M Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, 600113, India.
| |
Collapse
|
24
|
Xu HY, Zhang HX, Xiao Z, Qiao J, Li R. Regulation of anti-Müllerian hormone (AMH) in males and the associations of serum AMH with the disorders of male fertility. Asian J Androl 2020; 21:109-114. [PMID: 30381580 PMCID: PMC6413543 DOI: 10.4103/aja.aja_83_18] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Anti-Müllerian hormone (AMH) is a functional marker of fetal Sertoli cells. The germ cell number in adults depends on the number of Sertoli cells produced during perinatal development. Recently, AMH has received increasing attention in research of disorders related to male fertility. This paper reviews and summarizes the articles on the regulation of AMH in males and the serum levels of AMH in male fertility-related disorders. We have determined that follicle-stimulating hormone (FSH) promotes AMH transcription in the absence of androgen signaling. Testosterone inhibits the transcriptional activation of AMH. The undetectable levels of serum AMH and testosterone levels indicate a lack of functional testicular tissue, for example, that in patients with anorchia or severe Klinefelter syndrome suffering from impaired spermatogenesis. The normal serum testosterone level and undetectable AMH are highly suggestive of persistent Müllerian duct syndrome (PMDS), combined with clinical manifestations. The levels of both AMH and testosterone are always subnormal in patients with mixed disorders of sex development (DSD). Mixed DSD is an early-onset complete type of disorder with fetal hypogonadism resulting from the dysfunction of both Leydig and Sertoli cells. Serum AMH levels are varying in patients with male fertility-related disorders, including pubertal delay, severe congenital hypogonadotropic hypogonadism, nonobstructive azoospermia, Klinefelter syndrome, varicocele, McCune-Albright syndrome, and male senescence.
Collapse
Affiliation(s)
- Hui-Yu Xu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China.,Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing 100191, China.,National Clinical Research Center for Obstetrics and Gynecology, Beijing 100191, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China
| | - Hong-Xian Zhang
- Department of Urology, Peking University Third Hospital, Beijing 100191, China
| | - Zhen Xiao
- Department of Reproductive Endocrinology, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou 310014, China
| | - Jie Qiao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China.,Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing 100191, China.,National Clinical Research Center for Obstetrics and Gynecology, Beijing 100191, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China
| | - Rong Li
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China.,Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing 100191, China.,National Clinical Research Center for Obstetrics and Gynecology, Beijing 100191, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China
| |
Collapse
|
25
|
Vaughan D, Ritvos O, Mitchell R, Kretz O, Lalowski M, Amthor H, Chambers D, Matsakas A, Pasternack A, Collins-Hooper H, Ballesteros R, Huber TB, Denecke B, Widera D, Mukherjee A, Patel K. Inhibition of Activin/Myostatin signalling induces skeletal muscle hypertrophy but impairs mouse testicular development. Eur J Transl Myol 2020; 30:8737. [PMID: 32499882 PMCID: PMC7254437 DOI: 10.4081/ejtm.2019.8737] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 12/05/2019] [Indexed: 01/22/2023] Open
Abstract
Numerous approaches are being developed to promote post-natal muscle growth based on attenuating Myostatin/Activin signalling for clinical uses such as the treatment neuromuscular diseases, cancer cachexia and sarcopenia. However there have been concerns about the effects of inhibiting Activin on tissues other than skeletal muscle. We intraperitoneally injected mice with the Activin ligand trap, sActRIIB, in young, adult and a progeric mouse model. Treatment at any stage in the life of the mouse rapidly increased muscle mass. However at all stages of life the treatment decreased the weights of the testis. Not only were the testis smaller, but they contained fewer sperm compared to untreated mice. We found that the hypertrophic muscle phenotype was lost after the cessation of sActRIIB treatment but abnormal testis phenotype persisted. In summary, attenuation of Myostatin/Activin signalling inhibited testis development. Future use of molecules based on a similar mode of action to promote muscle growth should be carefully profiled for adverse side-effects on the testis. However the effectiveness of sActRIIB as a modulator of Activin function provides a possible therapeutic strategy to alleviate testicular seminoma development.
Collapse
Affiliation(s)
| | - Olli Ritvos
- Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
| | | | - Oliver Kretz
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maciej Lalowski
- Department of Biochemistry and Developmental Biology, HiLIFE, Meilahti Clinical Proteomics Core Facility, University of Helsinki, Helsinki, Finland
| | - Helge Amthor
- Versailles Saint-Quentin-en-Yvelines University, INSERM U1179, LIA BAHN CSM, Montigny-le-Bretonneux 78180, France
| | | | - Antonios Matsakas
- Molecular Physiology Laboratory, Centre for Atherothrombosis & Metabolic Disease, Hull York Medical School, Hull, UK
| | - Arja Pasternack
- Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
| | | | | | - Tobias B Huber
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | | - Ketan Patel
- School of Biological Sciences, University of Reading, UK
| |
Collapse
|
26
|
Edelsztein NY, Rey RA. Importance of the Androgen Receptor Signaling in Gene Transactivation and Transrepression for Pubertal Maturation of the Testis. Cells 2019; 8:E861. [PMID: 31404977 PMCID: PMC6721648 DOI: 10.3390/cells8080861] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/30/2019] [Accepted: 08/01/2019] [Indexed: 12/27/2022] Open
Abstract
Androgens are key for pubertal development of the mammalian testis, a phenomenon that is tightly linked to Sertoli cell maturation. In this review, we discuss how androgen signaling affects Sertoli cell function and morphology by concomitantly inhibiting some processes and promoting others that contribute jointly to the completion of spermatogenesis. We focus on the molecular mechanisms that underlie anti-Müllerian hormone (AMH) inhibition by androgens at puberty, as well as on the role androgens have on Sertoli cell tight junction formation and maintenance and, consequently, on its effect on proper germ cell differentiation and meiotic onset during spermatogenesis.
Collapse
Affiliation(s)
- Nadia Y Edelsztein
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE) - CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires C1425EFD, Argentina.
| | - Rodolfo A Rey
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE) - CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires C1425EFD, Argentina.
- Departamento de Biología Celular, Histología, Embriología y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina.
| |
Collapse
|
27
|
O'Shaughnessy PJ, Mitchell RT, Monteiro A, O'Hara L, Cruickshanks L, der Grinten HCV, Brown P, Abel M, Smith LB. Androgen receptor expression is required to ensure development of adult Leydig cells and to prevent development of steroidogenic cells with adrenal characteristics in the mouse testis. BMC DEVELOPMENTAL BIOLOGY 2019; 19:8. [PMID: 30995907 PMCID: PMC6472051 DOI: 10.1186/s12861-019-0189-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 03/29/2019] [Indexed: 01/10/2023]
Abstract
Background The interstitium of the mouse testis contains Leydig cells and a small number of steroidogenic cells with adrenal characteristics which may be derived from the fetal adrenal during development or may be a normal subset of the developing fetal Leydig cells. Currently it is not known what regulates development and/or proliferation of this sub-population of steroidogenic cells in the mouse testis. Androgen receptors (AR) are essential for normal testicular function and in this study we have examined the role of the AR in regulating interstitial cell development. Results Using a mouse model which lacks gonadotropins and AR (hpg.ARKO), stimulation of luteinising hormone receptors in vivo with human chorionic gonadotropin (hCG) caused a marked increase in adrenal cell transcripts/protein in a group of testicular interstitial cells. hCG also induced testicular transcripts associated with basic steroidogenic function in these mice but had no effect on adult Leydig cell-specific transcript levels. In hpg mice with functional AR, treatment with hCG induced Leydig cell-specific function and had no effect on adrenal transcript levels. Examination of mice with cell-specific AR deletion and knockdown of AR in a mouse Leydig cell line suggests that AR in the Leydig cells are likely to regulate these effects. Conclusions This study shows that in the mouse the androgen receptor is required both to prevent development of testicular cells with adrenal characteristics and to ensure development of an adult Leydig cell phenotype. Electronic supplementary material The online version of this article (10.1186/s12861-019-0189-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Peter J O'Shaughnessy
- College of Medical, Veterinary and Life Sciences, Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, G61 1QH, Glasgow, UK.
| | - Rod T Mitchell
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Ana Monteiro
- College of Medical, Veterinary and Life Sciences, Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, G61 1QH, Glasgow, UK
| | - 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.,Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK
| | - Lyndsey Cruickshanks
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Hedi Claahsen-van der Grinten
- Department of Paediatrics, Radboud Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Pamela Brown
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Margaret Abel
- Department of Human Anatomy and Genetics, University of Oxford, South Parks Rd, Oxford, OX1 3QX, 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.,School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW, 2308, Australia
| |
Collapse
|
28
|
Meroni SB, Galardo MN, Rindone G, Gorga A, Riera MF, Cigorraga SB. Molecular Mechanisms and Signaling Pathways Involved in Sertoli Cell Proliferation. Front Endocrinol (Lausanne) 2019; 10:224. [PMID: 31040821 PMCID: PMC6476933 DOI: 10.3389/fendo.2019.00224] [Citation(s) in RCA: 157] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 03/21/2019] [Indexed: 12/16/2022] Open
Abstract
Sertoli cells are somatic cells present in seminiferous tubules which have essential roles in regulating spermatogenesis. Considering that each Sertoli cell is able to support a limited number of germ cells, the final number of Sertoli cells reached during the proliferative period determines sperm production capacity. Only immature Sertoli cells, which have not established the blood-testis barrier, proliferate. A number of hormonal cues regulate Sertoli cell proliferation. Among them, FSH, the insulin family of growth factors, activin, and cytokines action must be highlighted. It has been demonstrated that cAMP/PKA, ERK1/2, PI3K/Akt, and mTORC1/p70SK6 pathways are the main signal transduction pathways involved in Sertoli cell proliferation. Additionally, c-Myc and hypoxia inducible factor are transcription factors which participate in the induction by FSH of various genes of relevance in cell cycle progression. Cessation of proliferation is a pre-requisite to Sertoli cell maturation accompanied by the establishment of the blood-testis barrier. With respect to this barrier, the participation of androgens, estrogens, thyroid hormones, retinoic acid and opioids has been reported. Additionally, two central enzymes that are involved in sensing cell energy status have been associated with the suppression of Sertoli cell proliferation, namely AMPK and Sirtuin 1 (SIRT1). Among the molecular mechanisms involved in the cessation of proliferation and in the maturation of Sertoli cells, it is worth mentioning the up-regulation of the cell cycle inhibitors p21Cip1, p27Kip, and p19INK4, and of the gap junction protein connexin 43. A decrease in Sertoli cell proliferation due to administration of certain therapeutic drugs and exposure to xenobiotic agents before puberty has been experimentally demonstrated. This review focuses on the hormones, locally produced factors, signal transduction pathways, and molecular mechanisms controlling Sertoli cell proliferation and maturation. The comprehension of how the final number of Sertoli cells in adulthood is established constitutes a pre-requisite to understand the underlying causes responsible for the progressive decrease in sperm production that has been observed during the last 50 years in humans.
Collapse
|
29
|
Câmara ML, Almeida TB, de Santi F, Rodrigues BM, Cerri PS, Beltrame FL, Sasso-Cerri E. Fluoxetine-induced androgenic failure impairs the seminiferous tubules integrity and increases ubiquitin carboxyl-terminal hydrolase L1 (UCHL1): Possible androgenic control of UCHL1 in germ cell death? Biomed Pharmacother 2018; 109:1126-1139. [PMID: 30551363 DOI: 10.1016/j.biopha.2018.10.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 09/28/2018] [Accepted: 10/09/2018] [Indexed: 12/21/2022] Open
Abstract
The selective serotonin reuptake inhibitor fluoxetine has been used for the treatment of depression. Although sexual disorders have been reported in male patients, few studies have demonstrated the fluoxetine effect on the reproductive histophysiology, and the target of this antidepressant in testes is unknown. We evaluated the impact of short-term treatment with fluoxetine on the adult rat testes, focusing on steroidogenesis by Leydig cells (LC) and androgen-dependent testicular parameters, including Sertoli cells (SC) and peritubular myoid cells (PMC). Since UCHL1 (ubiquitincarboxyl-terminal hydrolase L1) seems to control spermatogenesis, the immunoexpression of this hydrolase was also analyzed. Adult male rats received 20 mg/kg BW of fluoxetine (FG) or saline (CG) for eleven days. In historesin-embedded testis sections, the seminiferous tubule (ST) and epithelial (Ep) areas, and the LC nuclear diameter (LCnu) were measured. The number of abnormal ST, androgen-dependent ST, SC and PMC was quantified. Testicular β-tubulin levels and peritubular actin immunofluorescence were evaluated. Serum testosterone levels (STL) and steroidogenesis by 17β-HSD6 immunofluorescence were analyzed, and either UCHL1-immunolabeled or TUNEL-positive germ cells were quantified. In FG, abnormal ST frequency increased whereas ST and Ep areas, androgen-dependent ST number, LCnu, 17β-HSD6 activity and STL reduced significantly. TUNEL-positive PMC and SC was related to decreased number of these cells and reduction in peritubular actin and β-tubulin levels. In FG, uncommon UCHL1-immunoexpression was found in spermatocytes and spermatids, and the number of UCHL1-immunolabeled and TUNEL-positive germ cells increased in this group. These findings indicate that LC may be a fluoxetine target in testes, impairing PMC-SC integrity and disturbing spermatogenesis. The increase of UCHL1 in the damaged tubules associated with high incidence of cell death confirms that this hydrolase regulates germ cell death and may be controlled by androgens. The fertility in association with the androgenic status of patients treated with fluoxetine should be carefully evaluated.
Collapse
Affiliation(s)
- Marina L Câmara
- Dental School of São Paulo State University, Department of Morphology, Araraquara, Brazil
| | - Talita B Almeida
- Dental School of São Paulo State University, Department of Morphology, Araraquara, Brazil
| | - Fabiane de Santi
- Federal University of São Paulo, Department of Morphology and Genetics, São Paulo, Brazil
| | - Beatriz M Rodrigues
- Dental School of São Paulo State University, Department of Morphology, Araraquara, Brazil
| | - Paulo S Cerri
- Dental School of São Paulo State University, Department of Morphology, Araraquara, Brazil
| | - Flávia L Beltrame
- Federal University of São Paulo, Department of Morphology and Genetics, São Paulo, Brazil
| | - Estela Sasso-Cerri
- Dental School of São Paulo State University, Department of Morphology, Araraquara, Brazil.
| |
Collapse
|
30
|
Kojima K, Nakamura H, Komeya M, Yamanaka H, Makino Y, Okada Y, Akiyama H, Torikai N, Sato T, Fujii T, Kimura H, Ogawa T. Neonatal testis growth recreated in vitro by two-dimensional organ spreading. Biotechnol Bioeng 2018; 115:3030-3041. [PMID: 30144353 PMCID: PMC6283240 DOI: 10.1002/bit.26822] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 07/27/2018] [Accepted: 08/23/2018] [Indexed: 01/15/2023]
Abstract
Organ culture experiments can be hampered by central degeneration or necrosis due to the inadequate permeation of oxygen and nutrients, which deteriorates the function and growth of cultured tissues. In the current study, we aimed to overcome this limitation of organ culture through spreading the tissue two dimensionally on an agarose gel stand and molding into a disc shape by placing a ceiling of polydimethylsiloxane (PDMS) chip, which is highly oxygen permeable. By this, every part of the tissue can receive a sufficient supply of oxygen through PDMS as well as nutrients through the agarose gel below. This method not only prevented central necrosis of tissues, but also supported the tissue growth over time. In addition, such growth, as volume enlargement, could be easily measured. Under these conditions, we examined the effect of several factors on the growth of neonatal mouse testis, and found that follicle stimulating hormone (FSH) and insulin significantly promoted the growth. These results are in good agreement with previous in vivo reports. Notably, the growth achieved over 7 days in our in vitro system is almost comparable to, about 80% of, that observed in vivo. Thus, we successfully monitored the promotion of tissue growth beyond the limits of the conventional organ culture method. This extremely simple method could offer a unique platform to evaluate the growth as well as functional properties of organs, not only the testis but also others as well.
Collapse
Affiliation(s)
- Kazuaki Kojima
- Laboratory of Biopharmaceutical and Regenerative Sciences, Institute of Molecular Medicine and Life Science, Association of Medical Science, Yokohama City University, Yokohama, Japan
| | - Hiroko Nakamura
- Department of Mechanical Engineering, Tokai University, Hiratsuka, Japan
| | - Mitsuru Komeya
- Department of Urology, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Hiroyuki Yamanaka
- Department of Urology, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Yoshinori Makino
- Laboratory of Pathology and Development, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo, Japan
| | - Yuki Okada
- Laboratory of Pathology and Development, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo, Japan
| | - Haruhiko Akiyama
- Department of Orthopaedic Surgery, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Nobuhito Torikai
- Laboratory of Biopharmaceutical and Regenerative Sciences, Institute of Molecular Medicine and Life Science, Association of Medical Science, Yokohama City University, Yokohama, Japan
| | - Takuya Sato
- Laboratory of Biopharmaceutical and Regenerative Sciences, Institute of Molecular Medicine and Life Science, Association of Medical Science, Yokohama City University, Yokohama, Japan
| | - Teruo Fujii
- Institute of Industrial Science, University of Tokyo, Tokyo, Japan
| | - Hiroshi Kimura
- Department of Mechanical Engineering, Tokai University, Hiratsuka, Japan
| | - Takehiko Ogawa
- Laboratory of Biopharmaceutical and Regenerative Sciences, Institute of Molecular Medicine and Life Science, Association of Medical Science, Yokohama City University, Yokohama, Japan.,Department of Urology, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| |
Collapse
|
31
|
Li Y, Schang G, Wang Y, Zhou X, Levasseur A, Boyer A, Deng CX, Treier M, Boehm U, Boerboom D, Bernard DJ. Conditional Deletion of FOXL2 and SMAD4 in Gonadotropes of Adult Mice Causes Isolated FSH Deficiency. Endocrinology 2018; 159:2641-2655. [PMID: 29800110 PMCID: PMC6692885 DOI: 10.1210/en.2018-00100] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 05/15/2018] [Indexed: 12/24/2022]
Abstract
The glycoprotein FSH, a product of pituitary gonadotrope cells, regulates ovarian follicle development in females and spermatogenesis in males. FSH is a heterodimer of the common α gonadotropin subunit and the hormone-specific FSHβ subunit (a product of the Fshb gene). Using a conditional knockout approach (Cre-lox), we previously demonstrated that Fshb expression in mice depends on the transcription factors forkhead box L2 (FOXL2) and SMAD4. Deletion of Foxl2 or Smad4 alone led to FSH deficiency, female subfertility, and oligozoospermia in males. Simultaneous deletion of the two genes yielded a greater suppression of FSH and female sterility. The Cre-driver used previously was first active during embryonic development. Therefore, it is unclear whether FOXL2 and SMAD4 play important roles in the development or adult function of gonadotropes, or both. To address this question, we developed a tamoxifen-inducible Cre-driver line, which enabled Foxl2 and Smad4 gene deletions in gonadotropes of adult mice. After tamoxifen treatment, females with previously demonstrated fertility exhibited profound reductions in FSH levels, arrested ovarian follicle development, and sterility. FSH levels were comparably reduced in males 1 or 2 months after treatment; however, spermatogenesis was unaffected. These data indicate that (1) FOXL2 and SMAD4 are necessary to maintain FSH synthesis in gonadotrope cells of adult mice, (2) FSH is essential for female reproduction but appears to be unnecessary for the maintenance of spermatogenesis in adult male mice, and (3) the inducible Cre-driver line developed here provides a powerful tool to interrogate gene function in gonadotrope cells of adult mice.
Collapse
Affiliation(s)
- Yining Li
- Department of Pharmacology & Therapeutics, McGill University, Montréal, Québec, Canada
| | - Gauthier Schang
- Department of Pharmacology & Therapeutics, McGill University, Montréal, Québec, Canada
| | - Ying Wang
- Department of Pharmacology & Therapeutics, McGill University, Montréal, Québec, Canada
| | - Xiang Zhou
- Department of Pharmacology & Therapeutics, McGill University, Montréal, Québec, Canada
| | - Adrien Levasseur
- Département de Biomédecine Vétérinaire, Université de Montréal, St-Hyacinthe, Québec, Canada
| | - Alexandre Boyer
- Département de Biomédecine Vétérinaire, Université de Montréal, St-Hyacinthe, Québec, Canada
| | - Chu-Xia Deng
- Faculty of Health Sciences, University of Macau, Macau, China
| | - Mathias Treier
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch, Germany
- Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Ulrich Boehm
- Department of Experimental Pharmacology, Center for Molecular Signaling, Saarland University School of Medicine, Homburg, Germany
| | - Derek Boerboom
- Département de Biomédecine Vétérinaire, Université de Montréal, St-Hyacinthe, Québec, Canada
| | - Daniel J Bernard
- Department of Pharmacology & Therapeutics, McGill University, Montréal, Québec, Canada
- Correspondence: Daniel J. Bernard, PhD, Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir William Osler, Room 1315, Montréal, Quebec H3G 1Y6, Canada. E-mail:
| |
Collapse
|
32
|
Zhang X, Zhu Y, Tian Y, Yan H, Ren L, Shi W, Zhu J, Zhang T. The application of the improved 3D rat testicular cells co-culture model on the in vitro toxicity research of HZ1006. Drug Chem Toxicol 2018; 42:526-535. [PMID: 29681204 DOI: 10.1080/01480545.2018.1458237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
The aims of the present research are to further validate the application of the improved three-dimensional (3 D) rat testicular cell co-culture model to evaluate the effects of various reprotoxic chemicals on the function of the main somatic cells, as well as on spermatogonial cell differentiation and even spermatogenesis, and to investigate the specific toxicant mechanisms in testes treated with HZ1006, a hydroxamate-based a hydroxamate-based histone deacetylase inhibitor (HDACI). Based on the characteristics of HZ1006, the appropriate exposure duration (8, 16, or 24 days), dosage (0, 3.125, 6.25, 12.5, or 25 μM) and toxic endpoints suitable for detection were selected in the experiments. The results showed inhibition of cell proliferation, reduced testosterone levels, and decreased spermatogonial cell meiosis-specific gene expression, as well as decreased protein levels of androgen receptor (AR) and decreased expression of the AR target gene PSA, accompanied by inhibition of Hdac6 expression after HZ1006 exposure in the 3 D rat testicular cell co-culture model. These findings indicate that the improved 3 D rat testicular cell co-culture model we have established has the potential to become a new testicular toxicity test system that can be used to test toxic characteristics and mechanisms of new compounds and has good application prospects, although more research on the model is required.
Collapse
Affiliation(s)
- Xiaofang Zhang
- a Department of Hygienic Toxicology and Center for Evaluation of Drug Safety , Second Military Medical University , Shanghai , China
| | - Yuping Zhu
- a Department of Hygienic Toxicology and Center for Evaluation of Drug Safety , Second Military Medical University , Shanghai , China
| | - Yijun Tian
- a Department of Hygienic Toxicology and Center for Evaluation of Drug Safety , Second Military Medical University , Shanghai , China
| | - Han Yan
- b Shanghai Institute of Parenthood Research (National Evaluation Centre for the Toxicology of Fertility Regulating Drugs) , Shanghai , China
| | - Lijun Ren
- a Department of Hygienic Toxicology and Center for Evaluation of Drug Safety , Second Military Medical University , Shanghai , China
| | - Wenjing Shi
- a Department of Hygienic Toxicology and Center for Evaluation of Drug Safety , Second Military Medical University , Shanghai , China
| | - Jiangbo Zhu
- a Department of Hygienic Toxicology and Center for Evaluation of Drug Safety , Second Military Medical University , Shanghai , China
| | - Tianbao Zhang
- a Department of Hygienic Toxicology and Center for Evaluation of Drug Safety , Second Military Medical University , Shanghai , China
| |
Collapse
|
33
|
Nikpour F, Tayefi H, Mohammadnejad D, Akbarzadeh A. Adverse Effects of Vincristine Chemotherapy on Cell Changes in Seminiferous Tubules and Cetrorelix GnRH Antagonist
Inhibitory Effects in Mice. Asian Pac J Cancer Prev 2018; 19:683-687. [PMID: 29580040 PMCID: PMC5980841 DOI: 10.22034/apjcp.2018.19.3.683] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The present study was designed to determine changes in spermatogenesis in adult mice after an intraperitoneal injection of vincristine. The effect of a GnRH antagonist synchronous to chemotherapy, which might protect spermatogenesis by halting cell division in spermatogenic cells, was also investigated.
Collapse
Affiliation(s)
- Fatemeh Nikpour
- Department of Histological Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | | | | | | |
Collapse
|
34
|
Rondanino C, Maouche A, Dumont L, Oblette A, Rives N. Establishment, maintenance and functional integrity of the blood-testis barrier in organotypic cultures of fresh and frozen/thawed prepubertal mouse testes. Mol Hum Reprod 2018; 23:304-320. [PMID: 28333312 DOI: 10.1093/molehr/gax017] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 03/13/2017] [Indexed: 02/05/2023] Open
Abstract
STUDY QUESTION Can the spatio-temporal formation of an intact blood-testis barrier (BTB), which is essential for the progression of spermatogenesis, be reproduced in cultures of fresh or frozen/thawed prepubertal mouse testes? SUMMARY ANSWER Organotypic cultures allow the establishment and maintenance of major BTB components and the formation of a functional BTB in mouse testicular tissues. WHAT IS KNOWN ALREADY In vitro maturation of prepubertal testicular tissues is a promising approach to restore fertility in adult survivors of childhood cancer. Although gametes can be successfully obtained from prepubertal mouse testes in organotypic cultures, the spermatogenic yield remains low compared to in vivo controls. STUDY DESIGN, SIZE, DURATION Mouse testicular tissues were frozen using controlled slow freezing (CSF) or solid surface vitrification (SSV) procedures. A total of 158 testes (fresh n = 58, CSF n = 58 or SSV n = 42) from 6 to 7 days postpartum (dpp) mice were cultured at 34°C in basal medium (α-MEM, 10% KnockOut Serum Replacement, 5 μg/ml gentamicin) at a gas-liquid interphase (under 20% O2), with or without 10-6 M retinol, for 9, 16 and 30 days. In addition, 32 testes from 6-7, 15-16, 22-23 and 36-37 dpp mice were used as in vivo controls. PARTICIPANTS/MATERIALS, SETTING, METHODS The mRNA levels of BTB genes (Claudin 3, Claudin 11, Zonula occludens 1 and Connexin-43), germ cell-specific genes (Sal-like protein 4, Kit oncogene, Stimulated by retinoic acid gene 8, Synaptonemal complex protein 3, Transition protein 1 and Protamine 2), markers of Sertoli cell immaturity/maturity (anti-Mullerian hormone, androgen receptor, cyclin-dependent kinase inhibitor 1b) and the androgen-regulated gene Reproductive homeobox 5 (Rhox5) were measured by quantitative RT-PCR (RT-qPCR). The localization of BTB proteins in seminiferous tubules was studied by immunohistochemistry and spermatogenic progression was evaluated histologically. The integrity of the BTB was assessed using a biotin tracer. MAIN RESULTS AND THE ROLE OF CHANCE Modest differences in Claudin 11 (Cldn11), Zonula occludens 1 (Zo-1), Connexin-43 (Cx43) transcript levels and in the localization of the corresponding proteins were found between in vitro cultures of fresh or frozen/thawed testes and in vivo controls (P < 0.05). However, a 32-77-fold decrease in Claudin 3 (Cldn3) mRNA levels and a lack of CLDN3 immunolabelling in 36-44% of seminiferous tubules were observed in 30-day organotypic cultures (P < 0.05). Although Sertoli cell maturation and the completion of a full spermatogenic cycle were achieved after 30 days of culture, meiotic and postmeiotic progression was altered in cultured testicular tissues (P < 0.05). Moreover, an increased BTB permeability and a decreased expression of Rhox5 were observed at the end of the culture period in comparison with in vivo controls (P < 0.05). Completion of spermatogenesis occurred in vitro in seminiferous tubules with an intact BTB, and in those expressing or lacking CLDN3. LARGE SCALE DATA None. LIMITATIONS, REASONS FOR CAUTION Further studies will be needed to determine whether the expression of other BTB components is altered and to decipher the reason for lower Cldn3 and Rhox5 mRNA levels in organotypic cultures. WIDER IMPLICATIONS OF THE FINDINGS This work contributes to a better understanding of the molecular mechanisms occurring in in vitro matured prepubertal testes. The organotypic culture system will have to be developed further and optimized for human tissue, before potential clinical applications can be envisaged. STUDY FUNDING AND COMPETING INTEREST(S) This work was supported by Rouen University Hospital, Ligue contre le Cancer (to L.D.), and co-supported by European Union and Région Normandie (to A.O.). Europe gets involved in Normandie with European Régional Development Fund (ERDF). The authors declare that they have no conflict of interest.
Collapse
Affiliation(s)
- C Rondanino
- Normandie Univ, UNIROUEN, EA 4308 'Gametogenesis and Gamete Quality', Rouen University Hospital, Department of Reproductive Biology-CECOS, F 76000 Rouen, France
| | - A Maouche
- Normandie Univ, UNIROUEN, EA 4308 'Gametogenesis and Gamete Quality', Rouen University Hospital, Department of Reproductive Biology-CECOS, F 76000 Rouen, France
| | - L Dumont
- Normandie Univ, UNIROUEN, EA 4308 'Gametogenesis and Gamete Quality', Rouen University Hospital, Department of Reproductive Biology-CECOS, F 76000 Rouen, France
| | - A Oblette
- Normandie Univ, UNIROUEN, EA 4308 'Gametogenesis and Gamete Quality', Rouen University Hospital, Department of Reproductive Biology-CECOS, F 76000 Rouen, France
| | - N Rives
- Normandie Univ, UNIROUEN, EA 4308 'Gametogenesis and Gamete Quality', Rouen University Hospital, Department of Reproductive Biology-CECOS, F 76000 Rouen, France
| |
Collapse
|
35
|
Cavalari FC, da Rosa LA, Escott GM, Dourado T, de Castro AL, Kohek MBDF, Ribeiro MFM, Partata WA, de Fraga LS, Loss EDS. Epitestosterone- and testosterone-replacement in immature castrated rats changes main testicular developmental characteristics. Mol Cell Endocrinol 2018; 461:112-121. [PMID: 28870779 DOI: 10.1016/j.mce.2017.08.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 08/29/2017] [Accepted: 08/31/2017] [Indexed: 01/28/2023]
Abstract
Epitestosterone is the 17α-epimer of testosterone and has been described as an anti-androgen, since it inhibits the effects produced by testosterone and dihydrotestosterone via the nuclear androgen receptor (nAR). However, epitestosterone also displays an effect which is similar to the non-classical effect of testosterone, depolarizing the membrane potential of Sertoli cells and inducing a rapid Ca2+ uptake. This study aimed to investigate the effects of a treatment with epitestosterone on developmental parameters of immature rats. Animals were chemically castrated by using the gonadotropin-releasing hormone (GnRH) antagonist cetrorelix and then received a replacement of 7 days with epitestosterone or testosterone. Replacement with either epitestosterone or testosterone restored the anogenital distance (AGD) and testicular weight which had been reduced by chemical castration. The immunocontent of nAR and the nAR-immunoreactivity were reduced by epitestosterone treatment in the testis of both castrated and non-castrated animals. Furthermore, testosterone was unable of changing the membrane potential of Sertoli cells through its non-classical action in the group of animals castrated and replaced with epitestosterone. In conclusion, in relation to the level of protein expression of nAR epitestosterone acts as an anti-androgen. However, it acts in the same way as testosterone when genital development parameters are evaluated. Moreover, in castrated rats epitestosterone suppressed the non-classical response of testosterone, changing the pattern of testosterone signalling via a membrane mechanism in Sertoli cells.
Collapse
Affiliation(s)
- Fernanda Carvalho Cavalari
- Laboratório de Endocrinologia Experimental e Eletrofisiologia, Departamento de Fisiologia, PPG Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, Sala 337, Porto Alegre, RS, Brazil.
| | - Luciana Abreu da Rosa
- Laboratório de Endocrinologia Experimental e Eletrofisiologia, Departamento de Fisiologia, PPG Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, Sala 337, Porto Alegre, RS, Brazil.
| | - Gustavo Monteiro Escott
- Laboratório de Endocrinologia Experimental e Eletrofisiologia, Departamento de Fisiologia, PPG Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, Sala 337, Porto Alegre, RS, Brazil.
| | - Tadeu Dourado
- Laboratório de Endocrinologia Experimental e Eletrofisiologia, Departamento de Fisiologia, PPG Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, Sala 337, Porto Alegre, RS, Brazil.
| | - Alexandre Luz de Castro
- Centro Universitário Ritter dos Reis, UNIRITTER, Porto Alegre, RS, Brazil; Universidade Federal de Ciências da Saúde de Porto Alegre, UFCSPA, Porto Alegre, RS, Brazil.
| | | | - Maria Flávia Marques Ribeiro
- Laboratório de Interação Neuro-Humoral, Departamento de Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| | - Wania Aparecida Partata
- Laboratório de Neurobiologia Comparada, Departamento de Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| | - Luciano Stürmer de Fraga
- Laboratório de Endocrinologia Experimental e Eletrofisiologia, Departamento de Fisiologia, PPG Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, Sala 337, Porto Alegre, RS, Brazil.
| | - Eloísa da Silveira Loss
- Laboratório de Endocrinologia Experimental e Eletrofisiologia, Departamento de Fisiologia, PPG Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, Sala 337, Porto Alegre, RS, Brazil.
| |
Collapse
|
36
|
Jabarpour M, Tajik P. Evaluating the behavior of cultured sertoli cells in the presence and absence of spermatogonial stem cell. Stem Cell Investig 2018; 5:1. [PMID: 29430457 DOI: 10.21037/sci.2018.01.01] [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: 10/26/2017] [Accepted: 12/26/2017] [Indexed: 12/22/2022]
Abstract
Background The complex process of spermatogenesis is regulated by various factors. Several studies have been conducted to proliferate cells involved in the spermatogenesis process, in culture by used growth factors, different hormones and feeder cells. This study was conducted to evaluate the role of Sertoli cells on gene expression of fibroblast growth factor (FGF2) and glial cell derived neurotrophic factor (GDNF) after removal of spermatogonial stem cells (SSCs) from the culture medium. Methods Following isolation, bovine SSCs were co-cultured with Sertoli cells and follicular stimulating hormone (FSH) for 12 days. In the treatment group, SSCs were removed from the culture medium; in the control group no intervention was done in the culture. Colony formation of SSCs was evaluated by using an inverted microscope. Then, the expression of factors genes were assessed by quantitative RT-PCR. Data was analyzed by using paired-samples t-test. Results The results showed that removal of SSCs led to the increase in expression of GDNF and FGF2. These findings suggest that loss of SSCs population or decline in its population leads to changing in behavior of somatic cells which forming niche and consequently stimulates self-renewal and inhibits differentiation of SSCs. Conclusions The present study showed that removal of SSCs from the culture medium could be a model for damage to SSCs; the results revealed that niche cells respond to SSCs removal by upregulation of FGF2 and GDNF to stimulate self-renewal of SSCs and abrogation of differentiation.
Collapse
Affiliation(s)
- Masoome Jabarpour
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Parviz Tajik
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| |
Collapse
|
37
|
Tréfier A, Musnier A, Landomiel F, Bourquard T, Boulo T, Ayoub MA, León K, Bruneau G, Chevalier M, Durand G, Blache MC, Inoue A, Fontaine J, Gauthier C, Tesseraud S, Reiter E, Poupon A, Crépieux P. G protein-dependent signaling triggers a β-arrestin-scaffolded p70S6K/ rpS6 module that controls 5'TOP mRNA translation. FASEB J 2018; 32:1154-1169. [PMID: 29084767 DOI: 10.1096/fj.201700763r] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Many interaction partners of β-arrestins intervene in the control of mRNA translation. However, how β-arrestins regulate this cellular process has been poorly explored. In this study, we show that β-arrestins constitutively assemble a p70S6K/ribosomal protein S6 (rpS6) complex in HEK293 cells and in primary Sertoli cells of the testis. We demonstrate that this interaction is direct, and experimentally validate the interaction interface between β-arrestin 1 and p70S6K predicted by our docking algorithm. Like most GPCRs, the biological function of follicle-stimulating hormone receptor (FSHR) is transduced by G proteins and β-arrestins. Upon follicle-stimulating hormone (FSH) stimulation, activation of G protein-dependent signaling enhances p70S6K activity within the β-arrestin/p70S6K/rpS6 preassembled complex, which is not recruited to the FSHR. In agreement, FSH-induced rpS6 phosphorylation within the β-arrestin scaffold was decreased in cells depleted of Gαs. Integration of the cooperative action of β-arrestin and G proteins led to the translation of 5' oligopyrimidine track mRNA with high efficacy within minutes of FSH input. Hence, this work highlights new relationships between G proteins and β-arrestins when acting cooperatively on a common signaling pathway, contrasting with their previously shown parallel action on the ERK MAP kinase pathway. In addition, this study provides insights into how GPCR can exert trophic effects in the cell.-Tréfier, A., Musnier, A., Landomiel, F., Bourquard, T., Boulo, T., Ayoub, M. A., León, K., Bruneau, G., Chevalier, M., Durand, G., Blache, M.-C., Inoue, A., Fontaine, J., Gauthier, C., Tesseraud, S., Reiter, E., Poupon, A., Crépieux, P. G protein-dependent signaling triggers a β-arrestin-scaffolded p70S6K/ rpS6 module that controls 5'TOP mRNA translation.
Collapse
Affiliation(s)
- Aurélie Tréfier
- Biology and Bioinformatics of Signaling Systems (BIOS) Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique (INRA), Nouzilly, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 7247, Nouzilly, France.,Université François Rabelais, Tours, France.,Institut Français du Cheval et de l'Équitation (IFCE), Nouzilly, France
| | - Astrid Musnier
- Biology and Bioinformatics of Signaling Systems (BIOS) Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique (INRA), Nouzilly, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 7247, Nouzilly, France.,Université François Rabelais, Tours, France.,Institut Français du Cheval et de l'Équitation (IFCE), Nouzilly, France
| | - Flavie Landomiel
- Biology and Bioinformatics of Signaling Systems (BIOS) Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique (INRA), Nouzilly, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 7247, Nouzilly, France.,Université François Rabelais, Tours, France.,Institut Français du Cheval et de l'Équitation (IFCE), Nouzilly, France
| | - Thomas Bourquard
- Biology and Bioinformatics of Signaling Systems (BIOS) Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique (INRA), Nouzilly, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 7247, Nouzilly, France.,Université François Rabelais, Tours, France.,Institut Français du Cheval et de l'Équitation (IFCE), Nouzilly, France
| | - Thomas Boulo
- Biology and Bioinformatics of Signaling Systems (BIOS) Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique (INRA), Nouzilly, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 7247, Nouzilly, France.,Université François Rabelais, Tours, France.,Institut Français du Cheval et de l'Équitation (IFCE), Nouzilly, France
| | - Mohammed Akli Ayoub
- Biology and Bioinformatics of Signaling Systems (BIOS) Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique (INRA), Nouzilly, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 7247, Nouzilly, France.,Université François Rabelais, Tours, France.,Institut Français du Cheval et de l'Équitation (IFCE), Nouzilly, France.,Biology Department, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Kelly León
- Biology and Bioinformatics of Signaling Systems (BIOS) Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique (INRA), Nouzilly, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 7247, Nouzilly, France.,Université François Rabelais, Tours, France.,Institut Français du Cheval et de l'Équitation (IFCE), Nouzilly, France
| | - Gilles Bruneau
- Biology and Bioinformatics of Signaling Systems (BIOS) Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique (INRA), Nouzilly, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 7247, Nouzilly, France.,Université François Rabelais, Tours, France.,Institut Français du Cheval et de l'Équitation (IFCE), Nouzilly, France
| | - Manon Chevalier
- Biology and Bioinformatics of Signaling Systems (BIOS) Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique (INRA), Nouzilly, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 7247, Nouzilly, France.,Université François Rabelais, Tours, France.,Institut Français du Cheval et de l'Équitation (IFCE), Nouzilly, France
| | - Guillaume Durand
- Biology and Bioinformatics of Signaling Systems (BIOS) Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique (INRA), Nouzilly, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 7247, Nouzilly, France.,Université François Rabelais, Tours, France.,Institut Français du Cheval et de l'Équitation (IFCE), Nouzilly, France
| | - Marie-Claire Blache
- Plateau d'Imagerie Cellulaire (PIC), Unité Mixte de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique (INRA), Nouzilly, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 7247, Nouzilly, France.,Université François Rabelais, Tours, France.,Institut Français du Cheval et de l'Équitation (IFCE), Nouzilly, France
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan; and
| | - Joël Fontaine
- Biology and Bioinformatics of Signaling Systems (BIOS) Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique (INRA), Nouzilly, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 7247, Nouzilly, France.,Université François Rabelais, Tours, France.,Institut Français du Cheval et de l'Équitation (IFCE), Nouzilly, France
| | - Christophe Gauthier
- Biology and Bioinformatics of Signaling Systems (BIOS) Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique (INRA), Nouzilly, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 7247, Nouzilly, France.,Université François Rabelais, Tours, France.,Institut Français du Cheval et de l'Équitation (IFCE), Nouzilly, France
| | - Sophie Tesseraud
- Metabolism of Birds, Quality and Adaptation (MOQA) Group, Unité de Recherches 83, Unité de Recherches Avicoles, Institut National de la Recherche Agronomique (INRA), Nouzilly, France
| | - Eric Reiter
- Biology and Bioinformatics of Signaling Systems (BIOS) Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique (INRA), Nouzilly, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 7247, Nouzilly, France.,Université François Rabelais, Tours, France.,Institut Français du Cheval et de l'Équitation (IFCE), Nouzilly, France
| | - Anne Poupon
- Biology and Bioinformatics of Signaling Systems (BIOS) Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique (INRA), Nouzilly, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 7247, Nouzilly, France.,Université François Rabelais, Tours, France.,Institut Français du Cheval et de l'Équitation (IFCE), Nouzilly, France
| | - Pascale Crépieux
- Biology and Bioinformatics of Signaling Systems (BIOS) Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique (INRA), Nouzilly, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 7247, Nouzilly, France.,Université François Rabelais, Tours, France.,Institut Français du Cheval et de l'Équitation (IFCE), Nouzilly, France
| |
Collapse
|
38
|
Jabarpour M, Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.. Evaluation of the effect of follicular stimulating hormone on the in vitro bovine spermatogonial stem cells self-renewal: An experimental study. Int J Reprod Biomed 2017. [DOI: 10.29252/ijrm.15.12.795] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
39
|
Yu C, Li Y, Liu M, Gao M, Li C, Yan H, Li C, Sun L, Mo L, Wu C, Qi X, Ren J. Critical Role of Hepatic Cyp450s in the Testis-Specific Toxicity of (5R)-5-Hydroxytriptolide in C57BL/6 Mice. Front Pharmacol 2017; 8:832. [PMID: 29209210 PMCID: PMC5702336 DOI: 10.3389/fphar.2017.00832] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 10/31/2017] [Indexed: 12/17/2022] Open
Abstract
Low solubility, tissue accumulation, and toxicity are chief obstacles to developing triptolide derivatives, so a better understanding of the pharmacokinetics and toxicity of triptolide derivatives will help with these limitations. To address this, we studied pharmacokinetics and toxicity of (5R)-5-hydroxytriptolide (LLDT-8), a novel triptolide derivative immunosuppressant in a conditional knockout (KO) mouse model with liver-specific deletion of CYP450 reductase. Compared to wild type (WT) mice, after LLDT-8 treatment, KO mice suffered severe testicular toxicity (decreased testicular weight, spermatocytes apoptosis) unlike WT mice. Moreover, KO mice had greater LLDT-8 exposure as confirmed with elevated AUC and Cmax, increased drug half-life, and greater tissue distribution. γ-H2AX, a marker of meiosis process, its localization and protein level in testis showed a distinct meiosis block induced by LLDT-8. RNA polymerase II (Pol II), an essential factor for RNA storage and synapsis in spermatogenesis, decreased in testes of KO mice after LLDT-8 treatment. Germ-cell line based assays confirmed that LLDT-8 selectively inhibited Pol II in spermatocyte-like cells. Importantly, the analysis of androgen receptor (AR) related genes showed that LLDT-8 did not change AR-related signaling in testes. Thus, hepatic CYP450s were responsible for in vivo metabolism and clearance of LLDT-8 and aggravated testicular injury may be due to increased LLDT-8 exposure in testis and subsequent Pol II reduction.
Collapse
Affiliation(s)
- Cunzhi Yu
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yu Li
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Mingxia Liu
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Man Gao
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Chenggang Li
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Hong Yan
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Chunzhu Li
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Lihan Sun
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, China
| | - Liying Mo
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, China
| | - Chunyong Wu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, China
| | - Xinming Qi
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jin Ren
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
40
|
Recabarren SE, Recabarren M, Sandoval D, Carrasco A, Padmanabhan V, Rey R, Richter HG, Perez-Marin CC, Sir-Petermann T, Rojas-Garcia PP. Puberty arises with testicular alterations and defective AMH expression in rams prenatally exposed to testosterone. Domest Anim Endocrinol 2017; 61:100-107. [PMID: 28783504 DOI: 10.1016/j.domaniend.2017.06.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 06/15/2017] [Accepted: 06/16/2017] [Indexed: 11/20/2022]
Abstract
The male gonadal tissue can be a sensitive target to the reprogramming effects of testosterone (T) during prenatal development. We have demonstrated that male lambs born to dams receiving T during pregnancy-a model system to the polycystic ovary syndrome (PCOS)-show a decreased number of germ cells early in life, and when adult, a reduced amount of sperm and ejaculate volume. These findings are a key to put attention to the male offspring of women bearing PCOS, as they are exposed to increased levels of androgen during pregnancy which can reprogram their reproductive outcome. A possible origin of these defects can be a disruption in the expression of the anti-Müllerian hormone (AMH), due to its critical role in gonadal function at many postnatal stages. Therefore, we addressed the impact of prenatal T excess on the expression of AMH and factors related to its expression like AP2, SOX9, FSHR, and AR in the testicular tissue through real-time PCR during the peripubertal age. We also analyzed the testicular morphology and quantified the number of Sertoli cells and germ cells to evaluate any further defect in the testicle. Experiments were performed in rams at 24 wk of age, hence, prior puberty. The experimental animals (T-males) consisted of rams born to mothers receiving 30 mg testosterone twice a wk from Day 30 to 90 of pregnancy and then increased to 40 mg until Day 120 of pregnancy. The control males (C-males) were born to mothers receiving the vehicle of the hormone. We found a significant increase in the expression of the mRNA of AMH and SOX9, but not of the AP2, FHSR nor AR, in the T-males. Moreover, T-males showed a dramatic decrease in the number of germ cells, together with a decrease in the weight of their testicles. The findings of the present study show that before puberty, T-males are manifesting clear signs of disruption in the gonadal functions probably due to an alteration in the expression pattern of the AMH gene. The precise way by which T reprograms the expression of AMH gene remains to be established.
Collapse
Affiliation(s)
- S E Recabarren
- Laboratory of Animal Physiology and Endocrinology (FISENLAB), Faculty of Veterinary Sciences, University of Concepción, Chillán, Chile
| | - M Recabarren
- Laboratory of Animal Physiology and Endocrinology (FISENLAB), Faculty of Veterinary Sciences, University of Concepción, Chillán, Chile
| | - D Sandoval
- Laboratory of Animal Physiology and Endocrinology (FISENLAB), Faculty of Veterinary Sciences, University of Concepción, Chillán, Chile
| | - A Carrasco
- Laboratory of Animal Physiology and Endocrinology (FISENLAB), Faculty of Veterinary Sciences, University of Concepción, Chillán, Chile
| | - V Padmanabhan
- Departments of Pediatrics and the Reproductive Sciences Program, University of Michigan, Ann Arbor, Michigan, USA
| | - R Rey
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina; Departamento de Biología Celular, Histología, Embriología y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - H G Richter
- Laboratory of Developmental Chronobiology (LDC), Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - C C Perez-Marin
- Department of Animal Medicine and Surgery, Faculty of Veterinary Medicine, University of Cordoba, Cordoba, Spain
| | - T Sir-Petermann
- Laboratory of Endocrinology and Metabolism, Department of Internal Medicine, Western Faculty of Medicine, University of Chile, Santiago, Chile
| | - P P Rojas-Garcia
- Laboratory of Animal Physiology and Endocrinology (FISENLAB), Faculty of Veterinary Sciences, University of Concepción, Chillán, Chile.
| |
Collapse
|
41
|
Rebourcet D, Darbey A, Monteiro A, Soffientini U, Tsai YT, Handel I, Pitetti JL, Nef S, Smith LB, O'Shaughnessy PJ. Sertoli Cell Number Defines and Predicts Germ and Leydig Cell Population Sizes in the Adult Mouse Testis. Endocrinology 2017; 158:2955-2969. [PMID: 28911170 PMCID: PMC5659676 DOI: 10.1210/en.2017-00196] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 06/29/2017] [Indexed: 01/10/2023]
Abstract
Sertoli cells regulate differentiation and development of the testis and are essential for maintaining adult testis function. To model the effects of dysregulating Sertoli cell number during development or aging, we have used acute diphtheria toxin-mediated cell ablation to reduce Sertoli cell population size. Results show that the size of the Sertoli cell population that forms during development determines the number of germ cells and Leydig cells that will be present in the adult testis. Similarly, the number of germ cells and Leydig cells that can be maintained in the adult depends directly on the size of the adult Sertoli cell population. Finally, we have used linear modeling to generate predictive models of testis cell composition during development and in the adult based on the size of the Sertoli cell population. This study shows that at all ages the size of the Sertoli cell population is predictive of resulting testicular cell composition. A reduction in Sertoli cell number/proliferation at any age will therefore lead to a proportional decrease in germ cell and Leydig cell numbers, with likely consequential effects on fertility and health.
Collapse
Affiliation(s)
- Diane Rebourcet
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow G61 1QH, United Kingdom
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Annalucia Darbey
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Ana Monteiro
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow G61 1QH, United Kingdom
| | - Ugo Soffientini
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow G61 1QH, United Kingdom
| | - Yi Ting Tsai
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Ian Handel
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian EH25 9RG, United Kingdom
| | - Jean-Luc Pitetti
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva 4, Switzerland
| | - Serge Nef
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva 4, Switzerland
| | - Lee B Smith
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
- Faculty of Science, University of Newcastle, Callaghan, New South Wales 2308, Australia
| | - Peter J O'Shaughnessy
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow G61 1QH, United Kingdom
| |
Collapse
|
42
|
Lara NLM, França LR. Neonatal hypothyroidism does not increase Sertoli cell proliferation in iNOS−/− mice. Reproduction 2017; 154:13-22. [DOI: 10.1530/rep-17-0111] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 04/10/2017] [Accepted: 04/18/2017] [Indexed: 01/05/2023]
Abstract
Sertoli cell (SC) proliferation in mice occurs until two weeks after birth and is mainly regulated by FSH and thyroid hormones. Previous studies have shown that transient neonatal hypothyroidism in laboratory rodents is able to extend SC mitotic activity, leading ultimately to higher testis size and daily sperm production (DSP) in adult animals. Moreover, we have shown that due to higher SC proliferation and lower germ cell apoptosis, iNOS deficiency in mice also results in higher testis size and DSP. Although the cell size was smaller, the Leydig cells (LCs) number per testis also significantly increased in iNOS−/−mice. Our aims in the present study were to investigate if the combination of neonatal hypothyroidism and iNOS deficiency promotes additive effects in SC number, testis size and DSP. Hypothyroidism was induced in wild-type (WT) and iNOS−/−mice using 6-propyl-2-thiouracil (PTU) through the mother’s drinking water from 0 to 20 days of age, and were sacrificed at adulthood. Our results showed that, in contrast to the WT mice in which testis size, DSP and SC numbers increased significantly by 20, 40 and 70% respectively, after PTU treatment, no additive effects were observed for these parameters in treated iNOS−/−mice, as well as for LC. No alterations were observed in spermatogenesis in any group evaluated. Although we still do not have an explanation for these intriguing findings, we are currently investigating whether thyroid hormones influence iNOS levels and/or counterbalance physiological effects of iNOS deficiency in testis function and spermatogenesis.
Collapse
|
43
|
Yanai S, Hirano T, Omotehara T, Takada T, Yoneda N, Kubota N, Yamamoto A, Mantani Y, Yokoyama T, Kitagawa H, Hoshi N. Prenatal and early postnatal NOAEL-dose clothianidin exposure leads to a reduction of germ cells in juvenile male mice. J Vet Med Sci 2017; 79:1196-1203. [PMID: 28579575 PMCID: PMC5559363 DOI: 10.1292/jvms.17-0154] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Neonicotinoids are pesticides used worldwide. They bind to insect nicotinic acetylcholine receptors (nAChRs) with high affinity. We previously reported that clothianidin (CTD), one of the latest neonicotinoids, reduced
antioxidant expression and induced germ cell death in the adult testis of vertebrates. Here, we investigated the male reproductive toxicity of prenatal and early postnatal exposure to CTD, because it is likely that developmental
exposure more severely affects the testis compared to adults due to the absence of the blood-testis barrier. Pregnant C57BL/6 mice were given water gel blended with CTD (0, 10 or 50 mg/kg/day; no-observed-adverse-effect-level
[NOAEL for mice]: 47.2 mg/kg/day) between gestational day 1 and 14 days post-partum. We then examined the testes of male offspring at postnatal day 14. The testis weights and the numbers of germ cells per seminiferous tubule were
decreased in the CTD-50 group, and abnormal tubules containing no germ cells appeared. Nevertheless, the apoptotic cell number and proliferative activity were not significantly different between the control and CTD-exposed groups.
There were no significant differences in the androgen-related parameters, such as the Leydig cell volume per testis, the Sertoli cell number and the tubule diameter. The present study is the first demonstration that in
utero and lactational exposures to CTD at around the NOAEL for mice reduce the germ cell number, but our findings suggest that these exposures do not affect steroidogenesis in Leydig cells during prenatal or early
postnatal life.
Collapse
Affiliation(s)
- Shogo Yanai
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo 657-8501, Japan
| | - Tetsushi Hirano
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo 657-8501, Japan
| | - Takuya Omotehara
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo 657-8501, Japan
| | - Tadashi Takada
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo 657-8501, Japan
| | - Naoki Yoneda
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo 657-8501, Japan
| | - Naoto Kubota
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo 657-8501, Japan
| | - Anzu Yamamoto
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo 657-8501, Japan
| | - Youhei Mantani
- Laboratory of Histophysiology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo 657-8501, Japan
| | - Toshifumi Yokoyama
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo 657-8501, Japan
| | - Hiroshi Kitagawa
- Laboratory of Histophysiology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo 657-8501, Japan
| | - Nobuhiko Hoshi
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo 657-8501, Japan
| |
Collapse
|
44
|
Abstract
PURPOSE OF REVIEW To describe pubertal testicular growth in humans, changes in testicular cell populations that result in testicular growth, and the role of testosterone and gonadotrophins follicle-stimulating hormone (FSH) and luteinizing hormone (LH) in testicular growth. When human data were not available, studies in nonhuman primates and/or rodents were used as surrogates. RECENT FINDINGS Testicular growth in puberty follows a sigmoidal growth curve, with a large variation in timing of testicular growth and adult testicular volume. Testicular growth early in puberty is due to increase in Sertoli cell number and length of seminiferous tubules, whereas the largest and fastest growth results from the increase in the diameter of the seminiferous tubules first due to spermatogonial proliferation and then due to the expansion of meiotic and haploid germ cells. FSH stimulates Sertoli cell and spermatogonial proliferation, whereas LH/testosterone is mandatory to complete spermatogenesis. However, FSH and LH/testosterone work in synergy and are both needed for normal spermatogenesis. SUMMARY Testicular growth during puberty is rapid, and mostly due to germ cell expansion and growth in seminiferous tubule diameter triggered by androgens. Pre-treatment with FSH before the induction of puberty may improve the treatment of hypogonadotropic hypogonadism, but remains to be proven.
Collapse
Affiliation(s)
- Jaakko J Koskenniemi
- Institute of Biomedicine, Department of Physiology, University of Turku, and Department of Paediatrics, Turku University Hospital, Turku, Finland
| | | | | |
Collapse
|
45
|
Santi D, Spaggiari G, Casarini L, Fanelli F, Mezzullo M, Pagotto U, Granata ARM, Carani C, Simoni M. Central hypogonadism due to a giant, "silent" FSH-secreting, atypical pituitary adenoma: effects of adenoma dissection and short-term Leydig cell stimulation by luteinizing hormone (LH) and human chorionic gonadotropin (hCG). Aging Male 2017; 20:96-101. [PMID: 28067604 DOI: 10.1080/13685538.2016.1276161] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
We present a case report of an atypical giant pituitary adenoma secreting follicle-stimulating hormone (FSH). A 55-year-old patient presented for erectile dysfunction, loss of libido and fatigue. The biochemical evaluation showed very high FSH serum levels in the presence of central hypogonadism. Neither testicular enlargement nor increased sperm count was observed, thus a secretion of FSH with reduced biological activity was supposed. The histological examination after neuro-surgery showed an atypical pituitary adenoma with FSH-positive cells. Hypogonadism persisted and semen analyses impaired until azoospermia in conjunction with the reduction in FSH levels suggesting that, at least in part, this gonadotropin should be biologically active. Thus, we hypothesized a concomitant primary testicular insufficiency. The patient underwent short-term treatment trials with low doses of either recombinant luteinizing hormone (LH) or human chorionic gonadotropin (hCG) in three consecutive treatment schemes, showing an equal efficacy in stimulating testosterone (T) increase. This is the first case of atypical, giant FSH-secreting pituitary adenoma with high FSH serum levels without signs of testicular hyperstimulation, in presence of hypogonadism with plausible combined primary and secondary etiology. Hypophysectomized patients may represent a good model to assess both pharmacodynamics and effective dose of LH and hCG in the male.
Collapse
Affiliation(s)
- Daniele Santi
- a Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia , Modena , Italy
- b Unit of Endocrinology, Department of Medicine, Endocrinology, Metabolism and Geriatrics, Azienda USL of Modena , Modena , Italy
| | - Giorgia Spaggiari
- a Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia , Modena , Italy
- b Unit of Endocrinology, Department of Medicine, Endocrinology, Metabolism and Geriatrics, Azienda USL of Modena , Modena , Italy
| | - Livio Casarini
- a Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia , Modena , Italy
- c Center for Genomic Research, University of Modena and Reggio Emilia , Modena , Italy , and
| | - Flaminia Fanelli
- d Endocrinology Unit and Centre for Applied Biomedical Research, Department of Medical and Surgical Sciences, S. Orsola-Malpighi Hospital, Alma Mater University of Bologna , Bologna , Italy
| | - Marco Mezzullo
- d Endocrinology Unit and Centre for Applied Biomedical Research, Department of Medical and Surgical Sciences, S. Orsola-Malpighi Hospital, Alma Mater University of Bologna , Bologna , Italy
| | - Uberto Pagotto
- d Endocrinology Unit and Centre for Applied Biomedical Research, Department of Medical and Surgical Sciences, S. Orsola-Malpighi Hospital, Alma Mater University of Bologna , Bologna , Italy
| | - Antonio R M Granata
- a Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia , Modena , Italy
- b Unit of Endocrinology, Department of Medicine, Endocrinology, Metabolism and Geriatrics, Azienda USL of Modena , Modena , Italy
| | - Cesare Carani
- a Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia , Modena , Italy
| | - Manuela Simoni
- a Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia , Modena , Italy
- b Unit of Endocrinology, Department of Medicine, Endocrinology, Metabolism and Geriatrics, Azienda USL of Modena , Modena , Italy
- c Center for Genomic Research, University of Modena and Reggio Emilia , Modena , Italy , and
| |
Collapse
|
46
|
Pampanini V, Germani D, Puglianiello A, Stukenborg JB, Reda A, Savchuk I, Kjartansdóttir KR, Cianfarani S, Söder O. Impact of uteroplacental insufficiency on postnatal rat male gonad. J Endocrinol 2017; 232:247-257. [PMID: 27885054 PMCID: PMC5184772 DOI: 10.1530/joe-16-0418] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 11/24/2016] [Indexed: 11/30/2022]
Abstract
Prenatal events such as intrauterine growth restriction can affect gonadal development of the offspring and have an impact on reproductive health. To investigate the effects of intrauterine growth restriction induced by uterine artery ligation on the postnatal rat testis. Pregnant rats underwent uterine artery ligation at day 19 of gestation. Offspring were killed at 5, 20 and 40 days post-partum (dpp). At killing, one gonad was snap-frozen in liquid nitrogen and processed for RNA and steroid extraction. The other gonad was formalin-fixed for histology. Gene expression was analyzed by TaqMan Low-Density Array. Intratesticular testosterone, estradiol and serum gonadotrophins were measured. Thirty genes were dysregulated in intrauterine growth-restricted rats compared to controls, among which markers of Sertoli cell and Leydig cell function, cell metabolism and growth factors. Testis weights were significantly reduced at 5 and 20 dpp in intrauterine growth-restricted rats and caught-up by 40 dpp Accordingly, Sertoli cell number was significantly lower in 5 dpp intrauterine growth-restricted rats. At 20 dpp, intratesticular testosterone was significantly increased in intrauterine growth-restricted rats, whereas serum gonadotrophins were unchanged. IUGR altered the gene expression in the rat testes up to peripubertal age and reduced testis size and Sertoli cell number in neonatal age. Multiple mechanisms encompassing genetic changes and steroid production by the testis may be involved in the catch-up growth phase that restored testis size by 40 dpp Permanent consequences on organ function and gamete integrity cannot be excluded and deserve further investigations.
Collapse
Affiliation(s)
- Valentina Pampanini
- Department of Women's and Children's HealthPediatric Endocrinology Unit, Q2:08, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Daniela Germani
- Department of Systems MedicineTor Vergata University, Rome, Italy
| | | | - Jan-Bernd Stukenborg
- Department of Women's and Children's HealthPediatric Endocrinology Unit, Q2:08, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Ahmed Reda
- Department of Women's and Children's HealthPediatric Endocrinology Unit, Q2:08, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Iuliia Savchuk
- Department of Women's and Children's HealthPediatric Endocrinology Unit, Q2:08, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Kristín Rós Kjartansdóttir
- Department of Women's and Children's HealthPediatric Endocrinology Unit, Q2:08, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Stefano Cianfarani
- Department of Women's and Children's HealthPediatric Endocrinology Unit, Q2:08, Karolinska Institutet and University Hospital, Stockholm, Sweden
- Dipartimento Pediatrico Universitario Ospedaliero 'Bambino Gesù' Children's Hospital - Tor Vergata UniversityRome, Italy
| | - Olle Söder
- Department of Women's and Children's HealthPediatric Endocrinology Unit, Q2:08, Karolinska Institutet and University Hospital, Stockholm, Sweden
| |
Collapse
|
47
|
Schoeller EL, Clark DD, Dey S, Cao NV, Semaan SJ, Chao LW, Kauffman AS, Stowers L, Mellon PL. Bmal1 Is Required for Normal Reproductive Behaviors in Male Mice. Endocrinology 2016; 157:4914-4929. [PMID: 27704948 PMCID: PMC5133342 DOI: 10.1210/en.2016-1620] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Circadian rhythms synchronize physiological processes with the light-dark cycle and are regulated by a hierarchical system initiated in the suprachiasmatic nucleus, a hypothalamic region that receives direct photic input. The suprachiasmatic nucleus then entrains additional oscillators in the periphery. Circadian rhythms are maintained by a molecular transcriptional feedback loop, of which brain and muscle aryl hydrocarbon receptor nuclear translocator-like protein 1 (BMAL1) is a key member. Disruption of circadian rhythms by deletion of the BMAL1 gene (Bmal1 knockout [KO]) induces a variety of disease states, including infertility in males, due to unidentified mechanisms. We find that, despite normal sperm function, Bmal1 KO males fail to mate with receptive females, indicating a behavioral defect. Mating is dependent on pheromone detection, as are several other behaviors. We determined that Bmal1 KO males also fail to display aggression and avoidance of predator scent, despite intact main olfactory function. Moreover, the vomeronasal organ, a specialized pheromone-responsive organ, was also functionally intact, as determined by calcium imaging in response to urine pheromone stimulus. However, neural circuit tracing using c-FOS activation revealed that, although Bmal1 KO males displayed appropriate activation in the olfactory bulb and accessory olfactory bulb, the bed nucleus of the stria terminalis and the medial preoptic area (areas responsible for integration of copulatory behaviors) failed to activate highly in response to the female scent. This indicates that neural signaling in select behavioral centers is impaired in the absence of BMAL1, likely underlying Bmal1 KO male copulatory defects, demonstrating the importance of the BMAL1 protein in the maintenance of neural circuits that drive pheromone-mediated mating behaviors.
Collapse
Affiliation(s)
- Erica L Schoeller
- Department of Reproductive Medicine and the Center for Reproductive Science and Medicine (E.L.S., D.D.C., N.V.C., S.J.S., L.W.C., A.S.K., P.L.M.), University of California, San Diego, La Jolla, California 92093-0674; and Department of Molecular and Cellular Neuroscience (S.D., L.S.), The Scripps Research Institute, La Jolla, California 92037
| | - Daniel D Clark
- Department of Reproductive Medicine and the Center for Reproductive Science and Medicine (E.L.S., D.D.C., N.V.C., S.J.S., L.W.C., A.S.K., P.L.M.), University of California, San Diego, La Jolla, California 92093-0674; and Department of Molecular and Cellular Neuroscience (S.D., L.S.), The Scripps Research Institute, La Jolla, California 92037
| | - Sandeepa Dey
- Department of Reproductive Medicine and the Center for Reproductive Science and Medicine (E.L.S., D.D.C., N.V.C., S.J.S., L.W.C., A.S.K., P.L.M.), University of California, San Diego, La Jolla, California 92093-0674; and Department of Molecular and Cellular Neuroscience (S.D., L.S.), The Scripps Research Institute, La Jolla, California 92037
| | - Nathan V Cao
- Department of Reproductive Medicine and the Center for Reproductive Science and Medicine (E.L.S., D.D.C., N.V.C., S.J.S., L.W.C., A.S.K., P.L.M.), University of California, San Diego, La Jolla, California 92093-0674; and Department of Molecular and Cellular Neuroscience (S.D., L.S.), The Scripps Research Institute, La Jolla, California 92037
| | - Sheila J Semaan
- Department of Reproductive Medicine and the Center for Reproductive Science and Medicine (E.L.S., D.D.C., N.V.C., S.J.S., L.W.C., A.S.K., P.L.M.), University of California, San Diego, La Jolla, California 92093-0674; and Department of Molecular and Cellular Neuroscience (S.D., L.S.), The Scripps Research Institute, La Jolla, California 92037
| | - Ling W Chao
- Department of Reproductive Medicine and the Center for Reproductive Science and Medicine (E.L.S., D.D.C., N.V.C., S.J.S., L.W.C., A.S.K., P.L.M.), University of California, San Diego, La Jolla, California 92093-0674; and Department of Molecular and Cellular Neuroscience (S.D., L.S.), The Scripps Research Institute, La Jolla, California 92037
| | - Alexander S Kauffman
- Department of Reproductive Medicine and the Center for Reproductive Science and Medicine (E.L.S., D.D.C., N.V.C., S.J.S., L.W.C., A.S.K., P.L.M.), University of California, San Diego, La Jolla, California 92093-0674; and Department of Molecular and Cellular Neuroscience (S.D., L.S.), The Scripps Research Institute, La Jolla, California 92037
| | - Lisa Stowers
- Department of Reproductive Medicine and the Center for Reproductive Science and Medicine (E.L.S., D.D.C., N.V.C., S.J.S., L.W.C., A.S.K., P.L.M.), University of California, San Diego, La Jolla, California 92093-0674; and Department of Molecular and Cellular Neuroscience (S.D., L.S.), The Scripps Research Institute, La Jolla, California 92037
| | - Pamela L Mellon
- Department of Reproductive Medicine and the Center for Reproductive Science and Medicine (E.L.S., D.D.C., N.V.C., S.J.S., L.W.C., A.S.K., P.L.M.), University of California, San Diego, La Jolla, California 92093-0674; and Department of Molecular and Cellular Neuroscience (S.D., L.S.), The Scripps Research Institute, La Jolla, California 92037
| |
Collapse
|
48
|
Egbowon BF, Harris W, Arnott G, Mills CL, Hargreaves AJ. Sub-lethal concentrations of CdCl2 disrupt cell migration and cytoskeletal proteins in cultured mouse TM4 Sertoli cells. Toxicol In Vitro 2016; 32:154-65. [DOI: 10.1016/j.tiv.2015.12.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 11/29/2015] [Accepted: 12/23/2015] [Indexed: 11/30/2022]
|
49
|
Jiang S, Katz TA, Garee JP, DeMayo FJ, Lee AV, Oesterreich S. Scaffold attachment factor B2 (SAFB2)-null mice reveal non-redundant functions of SAFB2 compared with its paralog, SAFB1. Dis Model Mech 2015; 8:1121-7. [PMID: 26092125 PMCID: PMC4582101 DOI: 10.1242/dmm.019885] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 06/09/2015] [Indexed: 01/03/2023] Open
Abstract
Scaffold attachment factors SAFB1 and SAFB2 are multifunctional proteins that share >70% sequence similarity. SAFB1-knockout (SAFB1(-/-)) mice display a high degree of lethality, severe growth retardation, and infertility in male mice. To assess the in vivo role of SAFB2, and to identify unique functions of the two paralogs, we generated SAFB2(-/-) mice. In stark contrast to SAFB1(-/-), SAFB2(-/-) offspring were born at expected Mendelian ratios and did not show any obvious defects in growth or fertility. Generation of paralog-specific antibodies allowed extensive expression analysis of SAFB1 and SAFB2 in mouse tissues, showing high expression of both SAFB1 and SAFB2 in the immune system, and in hormonally controlled tissues, with especially high expression of SAFB2 in the male reproductive tract. Further analysis showed a significantly increased testis weight in SAFB2(-/-) mice, which was associated with an increased number of Sertoli cells. Our data suggest that this is at least in part caused by alterations in androgen-receptor function and expression upon deletion of SAFB2. Thus, despite a high degree of sequence similarity, SAFB1(-/-) and SAFB2(-/-) mice do not totally phenocopy each other. SAFB2(-/-) mice are viable, and do not show any major defects, and our data suggest a role for SAFB2 in the differentiation and activity of Sertoli cells that deserves further study.
Collapse
Affiliation(s)
- Shiming Jiang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Tiffany A Katz
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Jason P Garee
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Francesco J DeMayo
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Adrian V Lee
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Steffi Oesterreich
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| |
Collapse
|
50
|
Legacki E, Conley AJ, Nitta-Oda BJ, Berger T. Porcine Sertoli Cell Proliferation after Androgen Receptor Inactivation1. Biol Reprod 2015; 92:93. [DOI: 10.1095/biolreprod.114.125716] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 02/16/2015] [Indexed: 01/23/2023] Open
|