1
|
Zhu W, Cheng X, Zhang H, Li J, Li L, Wei H, Zhang S. Cholic acid inhibits ovarian steroid hormone synthesis and follicular development through farnesoid X receptor signaling in mice. Int J Biol Macromol 2025; 301:140458. [PMID: 39884637 DOI: 10.1016/j.ijbiomac.2025.140458] [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: 10/19/2024] [Revised: 01/23/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
This study investigated the effects of cholic acid (CA) on steroid hormone synthesis and follicular development in mouse ovaries and the regulatory mechanism of CA on the expression of steroidogenesis-related genes in granulosa cells. The mice were divided into control and CA groups, and serum and ovarian samples were collected after 1, 2, and 4 months of treatment, respectively. The results showed that CA treatment for 1, 2, and 4 months reduced ovarian weights, disrupted the estrous cycle, decreased the numbers of antral follicles and corpora lutea, and lowered the serum levels of progesterone and estradiol. Moreover, in the ovary, CA treatment upregulated the expression of farnesoid X receptor (FXR) and downregulated the expression of steroidogenesis-related genes, including StAR, CYP11A1, and HSD3B1. Mechanistically, FXR knockdown reversed the inhibitory effects of CA on steroidogenesis-related gene expression and cholesterol uptake in granulosa cells. In vitro follicle culture experiments further confirmed that CA suppressed follicle development, decreased the mRNA expression of steroidogenesis-related genes, and reduced progesterone and estradiol secretion. Collectively, our results demonstrated that CA inhibited follicular development and steroid hormone synthesis through FXR signaling.
Collapse
Affiliation(s)
- Wenjun Zhu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Xiaochan Cheng
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Hengyu Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Jiahao Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Li Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
| | - Hengxi Wei
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
| | - Shouquan Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
2
|
Qiu X, Li W, Li X, Wu B, Dai M, Xia Y, Zhang G, Bian Y, Chen J, Wu K, Lu Y, Tang M, Lin H, Shang J. Discovery of Fluorescent Probe ABDS-2 for Farnesoid X Receptor Modulator Characterization and Cell-Based Imaging. Anal Chem 2025; 97:2019-2027. [PMID: 39841563 DOI: 10.1021/acs.analchem.4c03743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
The farnesoid X receptor (FXR) regulates key physiological processes, such as bile acid homeostasis and lipid metabolism, making it an important target for drug discovery. However, the overactivation of FXR often leads to adverse effects. This study presents the development of a novel fluorescent probe utilizing the computer-aided drug design (CADD) approach to optimize linkers between more potent warhead and FITC fluorescent groups. The probes were designed and assessed via molecular dynamics simulations, and four were selected for synthesis to be evaluated in in vitro biochemical assays. Among these, ABDS-2 exhibited high sensitivity and stability, which demonstrated satisfactory validation in high-throughput screening assays. Furthermore, ABDS-2 facilitated real-time bioimaging to monitor FXR homeostasis at the cellular level, providing spatially resolved insights into molecular interactions critical for cellular function studies. This research underscores the efficiency of CADD in probe design and positions ABDS-2 as a valuable chemical tool for in vitro assays and cellular-level bioimaging.
Collapse
Affiliation(s)
- Xianjie Qiu
- School of Basic Medical Sciences, Guangzhou Νational Laboratory, Guangzhou Medical University, Guangzhou 511436, China
| | - Wenqi Li
- School of Basic Medical Sciences, Guangzhou Νational Laboratory, Guangzhou Medical University, Guangzhou 511436, China
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiaoqin Li
- School of Basic Medical Sciences, Guangzhou Νational Laboratory, Guangzhou Medical University, Guangzhou 511436, China
| | - Bin Wu
- School of Basic Medical Sciences, Guangzhou Νational Laboratory, Guangzhou Medical University, Guangzhou 511436, China
- Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Minxian Dai
- School of Basic Medical Sciences, Guangzhou Νational Laboratory, Guangzhou Medical University, Guangzhou 511436, China
| | - Yi Xia
- School of Basic Medical Sciences, Guangzhou Νational Laboratory, Guangzhou Medical University, Guangzhou 511436, China
| | - Gong Zhang
- Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Yizhou Bian
- Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Jiayi Chen
- School of Basic Medical Sciences, Guangzhou Νational Laboratory, Guangzhou Medical University, Guangzhou 511436, China
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Kunzhong Wu
- School of Basic Medical Sciences, Guangzhou Νational Laboratory, Guangzhou Medical University, Guangzhou 511436, China
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yongzhi Lu
- School of Basic Medical Sciences, Guangzhou Νational Laboratory, Guangzhou Medical University, Guangzhou 511436, China
| | - Miru Tang
- School of Basic Medical Sciences, Guangzhou Νational Laboratory, Guangzhou Medical University, Guangzhou 511436, China
| | - Hua Lin
- Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Jinsai Shang
- School of Basic Medical Sciences, Guangzhou Νational Laboratory, Guangzhou Medical University, Guangzhou 511436, China
| |
Collapse
|
3
|
Gao X, Ke L, Wang L, Zheng S, Liu X, Hu W, Tong G, Li Z, Hu G. Low-temperature-induced disruption of reproductive axis and sperm vitality via stress axis in Monopterus albus. Gen Comp Endocrinol 2024; 359:114617. [PMID: 39368755 DOI: 10.1016/j.ygcen.2024.114617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 06/13/2024] [Accepted: 10/02/2024] [Indexed: 10/07/2024]
Abstract
The ricefield eel (Monopterus albus) is inherently timid and highly sensitive to stress. Our previous studies have shown that low-temperature weather could significantly affect the sperm vitality of ricefield eels. This study aims to investigate the regulatory mechanism of low-temperature effects on testicular function and sperm vitality in ricefield eels. The ricefield eels were initially reared at low (10 °C) and normal (25 °C) temperatures for 24 h. Low temperatures were found to induce the expression of pituitary pro-opiomelanocortin (POMC) and testes insulin-like growth factor-binding protein 1 (IGFBP1) mRNA expression, suggesting that the reduction in sperm vitality could be attributed to the activation of the stress axis. Moreover, the results indicated a significant decrease in sperm occupancy and count in the testes, along with a reduced percentage of motile sperm. Subsequent transcriptome analysis showed substantial inhibition of reproductive hormone genes (gnrh1, lh, and fsh) in the brain and pituitary, and downregulation of meiosis-related genes (dmc1, rec8, and sycp3) in the testes. These findings suggest that low temperatures might disrupt testicular development and spermatogenesis by inhibiting the reproductive axis. Metabolomics analysis then demonstrated a significant reduction in the levels of metabolites related to glycolysis, fatty acid metabolism, and the tricarboxylic acid (TCA) cycle in the testes after low-temperature treatment. Interestingly, the expression of zona pellucida sperm-binding proteins 3 and 4 (ZP3 and ZP4), which may affect sperm vitality and spermatogenesis, was significantly induced by low temperatures in the testes. In conclusion, these findings suggested that low temperatures might affect testicular function and sperm vitality by simultaneously activating the stress axis and inhibiting the reproductive axis and energy metabolism in the testes.
Collapse
Affiliation(s)
- Xiaowen Gao
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Liang Ke
- Hubei Provincial Rice Eel Industry Research Institute, Xiantao City 441409, China; Hubei Provincial Rice Eel Industry Group Co., Xiantao City 441409, China
| | - Linlin Wang
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Shuo Zheng
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiangjiang Liu
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Wenhao Hu
- Hubei Provincial Rice Eel Industry Group Co., Xiantao City 441409, China
| | - Guobing Tong
- Hong Yuan Ze Aquaculture Specialized Cooperative, China
| | - Zhong Li
- Hubei Provincial Rice Eel Industry Research Institute, Xiantao City 441409, China.
| | - Guangfu Hu
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China; Hubei Provincial Rice Eel Industry Research Institute, Xiantao City 441409, China.
| |
Collapse
|
4
|
Monrose M, Holota H, Martinez G, Damon-Soubeyrand C, Thirouard L, Martinot E, Battistelli E, de Haze A, Bravard S, Tamisier C, Caira F, Coutton C, Barbotin AL, Boursier A, Lakhal L, Beaudoin C, Volle DH. Constitutive Androstane Receptor Regulates Germ Cell Homeostasis, Sperm Quality, and Male Fertility via Akt-Foxo1 Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402082. [PMID: 39318179 DOI: 10.1002/advs.202402082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 07/02/2024] [Indexed: 09/26/2024]
Abstract
Male sexual function can be disrupted by exposure to exogenous compounds that cause testicular physiological alterations. The constitutive androstane receptor (Car) is a receptor for both endobiotics and xenobiotics involved in detoxification. However, its role in male fertility, particularly in regard to the reprotoxic effects of environmental pollutants, remains unclear. This study aims to investigate the role of the Car signaling pathway in male fertility. In vivo, in vitro, and pharmacological approaches are utilized in wild-type and Car-deficient mouse models. The results indicate that Car inhibition impaired male fertility due to altered sperm quality, specifically histone retention, which is correlated with an increased percentage of dying offspring in utero. The data highlighted interactions among Car, Akt, Foxo1, and histone acetylation. This study demonstrates that Car is crucial in germ cell homeostasis and male fertility. Further research on the Car signaling pathway is necessary to reveal unidentified causes of altered fertility and understand the harmful impact of environmental molecules on male fertility and offspring health.
Collapse
Affiliation(s)
- Mélusine Monrose
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
| | - Hélène Holota
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
| | - Guillaume Martinez
- CHU Grenoble Alpes, UM de Génétique Chromosomique, Grenoble, F-38000, France
- Team Genetics Epigenetics and Therapies of Infertility, Institute for Advanced Biosciences, University Grenoble Alpes, INSERM U1209, CNRS UMR 5309, Grenoble, F-38000, France
| | - Christelle Damon-Soubeyrand
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Plateform Anipath, Clermont-Ferrand, F-63001, France
| | - Laura Thirouard
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
| | - Emmanuelle Martinot
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
| | - Edwige Battistelli
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
| | - Angélique de Haze
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
| | - Stéphanie Bravard
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Plateform Anipath, Clermont-Ferrand, F-63001, France
| | - Christelle Tamisier
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
| | - Françoise Caira
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
| | - Charles Coutton
- CHU Grenoble Alpes, UM de Génétique Chromosomique, Grenoble, F-38000, France
- Team Genetics Epigenetics and Therapies of Infertility, Institute for Advanced Biosciences, University Grenoble Alpes, INSERM U1209, CNRS UMR 5309, Grenoble, F-38000, France
| | - Anne-Laure Barbotin
- CHU Lille, Institut de Biologie de la Reproduction-Spermiologie-CECOS, Lille, F-59000, France
- Inserm UMR-S 1172, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille, F-59000, France
| | - Angèle Boursier
- CHU Lille, Institut de Biologie de la Reproduction-Spermiologie-CECOS, Lille, F-59000, France
- Inserm UMR-S 1172, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille, F-59000, France
| | - Laila Lakhal
- INRAe UMR1331, ToxAlim, University of Toulouse, Toulouse, F-31027, France
| | - Claude Beaudoin
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
| | - David H Volle
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
| |
Collapse
|
5
|
Zou C, Wang W, Shu C, Liang S, Zou Y, Wang L, Wu Z, Liu Y, You F. Expression characteristics of Hsd3b7 in the gonads of Paralichthys olivaceus. Comp Biochem Physiol B Biochem Mol Biol 2023; 266:110848. [PMID: 36933762 DOI: 10.1016/j.cbpb.2023.110848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 02/20/2023] [Accepted: 03/08/2023] [Indexed: 03/18/2023]
Abstract
Steroidogenesis is an important biological process for gonadal differentiation and development. In mammals, 3β-hydroxysteroid dehydrogenase 7 (HSD3B7) could convert 3β-hydroxy of 7α-hydroxycholesterol into a ketone and form 7α-hydroxy-4-cholesten-3-one, which may affect steroidogenesis. However, in fish, the study of Hsd3b7 is still lacking. In this study, Hsd3b7 was identified in the olive flounder Paralichthys olivaceus, an important mariculture fish. According to bioinformatics analysis, Hsd3b7 belongs to a Rossmann-fold NAD(P)(+)-binding protein and can interact in a predictable manner with Hsd17b2, -3, and - 4, which play a role in steroidogenesis. In the adult flounder, Hsd3b7 was expressed in various tissues, at particularly high level in male muscle. The expression levels of Hsd3b7 at gonadal development stages I-V initially increased and then decreased, with an inflection point in the ovary at stage III and in the testis at stage IV. At stage III, the expression level of Hsd3b7 was significantly higher in the ovary than in the testis (P < 0.01). The results of in situ hybridization (ISH) revealed that it was mainly expressed in oocytes of phases I-IV or around oocytes of phases IV-V in the ovaries and around spermatid lobules at stages IV-V in the testes. Three regulatory sites of SRY-box transcription factor 9 (Sox9), a transcription factor involved in steroidogenesis and gonadal differentiation, were predicted in the promoter of Hsd3b7. After intraperitoneal injection with the recombination flounder Sox9a, the expression of Hsd3b7 was significantly up-regulated (P < 0.01). During the flounder gonadal differentiation, 17β-estradiol (E2, 5 μg/g feed) and 17α-methyltestosterone (T, 5 μg/g feed) were used to obtain the phenotypic female or male flounder, and the results showed that in the E2 group, Hsd3b7 expression was highest at 2 cm TL, the primordial gonad stage, which was significantly higher than that at 12 cm TL (P < 0.05). In the T group, Hsd3b7 expression level was also highest at 2 cm TL and significantly higher than at 10 and 12 cm TL (P < 0.05). Moreover, Hsd3b7 was detected to be localized mainly around oogonia and spermatogonia during the differentiated gonads with ISH. These findings first introduce the expression characteristics of Hsd3b7 and the effect of Sox9a on its expression, which contribute to our understanding of the function of Hsd3b7 in fish gonads.
Collapse
Affiliation(s)
- Congcong Zou
- Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, PR China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Wenxiang Wang
- Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, PR China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Chang Shu
- Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, PR China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Shaoshuai Liang
- Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, PR China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237, PR China
| | - Yuxia Zou
- Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, PR China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237, PR China
| | - Lijuan Wang
- Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, PR China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237, PR China
| | - Zhihao Wu
- Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, PR China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237, PR China
| | - Yan Liu
- Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, PR China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237, PR China
| | - Feng You
- Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, PR China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237, PR China.
| |
Collapse
|
6
|
Serra L, Bourdon G, Estienne A, Fréville M, Ramé C, Chevaleyre C, Didier P, Chahnamian M, Ganier P, Pinault F, Froment P, Dupont J. Triazole pesticides exposure impaired steroidogenesis associated to an increase in AHR and CAR expression in testis and altered sperm parameters in chicken. Toxicol Rep 2023; 10:409-427. [PMID: 37025555 PMCID: PMC10070196 DOI: 10.1016/j.toxrep.2023.03.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023] Open
Abstract
Since several decades, we observe the decline of various bird populations that could be partly linked to the agricultural intensification and the use of large amount of pesticides. Even if triazoles compounds are the most widely used fungicides, their effects on the reproductive parameters in birds are not clearly known. In the present study, we investigated the in vitro effects of 8 triazoles compounds alone (propiconazole (PP, from 0 to 10 µM), prothioconazole (PT), epoxiconazole (Epox), tetraconazole (TT), tebuconazole (TB), difenoconazole (Dif), cyproconazole (Cypro), metconazole (MC) (from 0 to 1 mM)) on the male chicken reproductive functions by using testis explants, primary Sertoli cells and sperm samples. In testis, all triazoles at the higher concentrations for 48 h inhibited lactate and testosterone secretion mostly in association with reduced expression of HSD3B and/or STAR mRNA levels. These data were also associated with increased expression of the nuclear receptors Aryl Hydrocarbon Receptor (AHR) and Constitutive Androstane Receptor (CAR) mRNA levels in testis and for all triazoles except for PP a reduction in Sertoli cell viability. When focusing on the sperm parameters, we demonstrated that most of the triazoles (MC, Epox, Dif, TB, TT and Cypro) at 0.1 or 1 mM for either 2, 12 or 24 min of exposure decreased sperm motility and velocity and increased the percentage of spermatozoa abnormal morphology. At the opposite, PP increased sperm motility in a dose dependent manner after 2 min of exposure whereas no significant effect was observed in response to PT whatever the dose and the time of exposure. Moreover, these effects were associated with an increase in the production of reactive oxygen species in spermatozoa. Taken together, most of the triazoles compounds impair testis steroidogenesis and semen parameters potentially through an increase in AHR and CAR expression and in oxidative stress, respectively. Data Availability Statement All the data will be available.
Collapse
|
7
|
Identification of the Role of TGR5 in the Regulation of Leydig Cell Homeostasis. Int J Mol Sci 2022; 23:ijms232315398. [PMID: 36499726 PMCID: PMC9738292 DOI: 10.3390/ijms232315398] [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: 09/30/2022] [Revised: 11/25/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Understanding the regulation of the testicular endocrine function leading to testosterone production is a major objective as the alteration of endocrine function is associated with the development of many diseases such as infertility. In the last decades, it has been demonstrated that several endogenous molecules regulate the steroidogenic pathway. Among them, bile acids have recently emerged as local regulators of testicular physiology and particularly endocrine function. Bile acids act through the nuclear receptor FXRα (Farnesoid-X-receptor alpha; NR1H4) and the G-protein-coupled bile acid receptor (GPBAR-1; TGR5). While FXRα has been demonstrated to regulate testosterone synthesis within Leydig cells, no data are available regarding TGR5. Here, we investigated the potential role of TGR5 within Leydig cells using cell culture approaches combined with pharmacological exposure to the TGR5 agonist INT-777. The data show that activation of TGR5 results in a decrease in testosterone levels. TGR5 acts through the PKA pathway to regulate steroidogenesis. In addition, our data show that TGR5 activation leads to an increase in cholesterol ester levels. This suggests that altered lipid homeostasis may be a mechanism explaining the TGR5-induced decrease in testosterone levels. In conclusion, the present work highlights the impact of the TGR5 signaling pathway on testosterone production and reinforces the links between bile acid signaling pathways and the testicular endocrine function. The testicular bile acid pathways need to be further explored to increase our knowledge of pathologies associated with impaired testicular endocrine function, such as fertility disorders.
Collapse
|
8
|
Wang Z, Yu J, Zhong S, Fan Z, Wang F, Ji C, Wang Y, Lei C, Dang R, Zhao F. Metabolomic profiling of Dezhou donkey seminal plasma related to freezability. Reprod Domest Anim 2022; 57:1165-1175. [PMID: 35713115 DOI: 10.1111/rda.14187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/14/2022] [Accepted: 06/15/2022] [Indexed: 12/12/2022]
Abstract
Donkeys are indispensable livestock in China because they have transport function and medicinal value. With the popularization of artificial insemination on donkeys, semen cryopreservation technology has gradually become a research hotspot. Seminal plasma is a necessary medium for transporting sperm and provides energy and nutrition for sperm. Seminal plasma metabolites play an important role in the process of sperm freezing, and also have an important impact on sperm motility and fertilization rate after freezing and thawing. In this study, liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis was used to compare the metabolic characteristics of seminal plasma of high freezability (HF) and low freezability (LF) male donkeys. We identified 672 metabolites from donkey seminal plasma, of which 33 metabolites were significantly different between the two groups. Metabolites were identified and categorized according to their major chemical classes, including homogeneous non-metal compounds, nucleosides, nucleotides, and analogues, organosulphur compounds, phenylpropanoids and polyketide, organoheterocyclic compounds, organic oxygen compounds, benzenoids, organic acids and derivatives, lipids and lipid-like molecules, organooxygen compounds, alkaloids and derivatives, organic nitrogen compounds. The results showed that the contents of phosphatidylcholine, piceatannol and enkephalin in donkey semen of HF group were significantly higher than those of LF group (p < .05), while the contents of taurocholic and lysophosphatidic acid were significantly lower than those of LF group (p < .05). The different metabolites were mainly related to sperm biological pathway response and oxidative stress. These metabolites may be considered as candidate biomarkers for different fertility in jacks.
Collapse
Affiliation(s)
- Zhaofei Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, Shaanxi Province, China
| | - Jie Yu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, Shaanxi Province, China.,Dong-E-E-Jiao Co. Ltd., National Engineering Research Center for Gelatin-Based Traditional Chinese Medicine, Done-E Country, China
| | - Shuai Zhong
- College of Pharmacy, Heze University, Heze, China
| | - Zhaobin Fan
- College of Pharmacy, Heze University, Heze, China
| | - Fuwen Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, Shaanxi Province, China
| | - Chuanliang Ji
- Dong-E-E-Jiao Co. Ltd., National Engineering Research Center for Gelatin-Based Traditional Chinese Medicine, Done-E Country, China
| | - Yantao Wang
- Dong-E-E-Jiao Co. Ltd., National Engineering Research Center for Gelatin-Based Traditional Chinese Medicine, Done-E Country, China
| | - Chuzhao Lei
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, Shaanxi Province, China
| | - Ruihua Dang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, Shaanxi Province, China
| | - Fuwei Zhao
- College of Pharmacy, Heze University, Heze, China
| |
Collapse
|
9
|
Thirouard L, Holota H, Monrose M, Garcia M, de Haze A, Damon‐Soubeyrand C, Renaud Y, Saru J, Perino A, Schoonjans K, Beaudoin C, Volle DH. Identification of a Crosstalk among TGR5, GLIS2, and TP53 Signaling Pathways in the Control of Undifferentiated Germ Cell Homeostasis and Chemoresistance. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200626. [PMID: 35435331 PMCID: PMC9189661 DOI: 10.1002/advs.202200626] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/09/2022] [Indexed: 06/14/2023]
Abstract
Spermatogonial stem cells regenerate and maintain spermatogenesis throughout life, making testis a good model for studying stem cell biology. The effects of chemotherapy on fertility have been well-documented previously. This study investigates how busulfan, an alkylating agent that is often used for chemotherapeutic purposes, affects male fertility. Specifically, the role of the TGR5 pathway is investigated on spermatogonia homeostasis using in vivo, in vitro, and pharmacological methods. In vivo studies are performed using wild-type and Tgr5-deficient mouse models. The results clearly show that Tgr5 deficiency can facilitate restoration of the spermatogonia homeostasis and allow faster resurgence of germ cell lineage after exposure to busulfan. TGR5 modulates the expression of key genes of undifferentiated spermatogonia such as Gfra1 and Fgfr2. At the molecular level, the present data highlight molecular mechanisms underlying the interactions among the TGR5, GLIS2, and TP53 pathways in spermatogonia associated with germ cell apoptosis following busulfan exposure. This study makes a significant contribution to the literature because it shows that TGR5 plays key role on undifferentiated germ cell homeostasis and that modulating the TGR5 signaling pathway could be used as a potential therapeutic tool for fertility disorders.
Collapse
Affiliation(s)
- Laura Thirouard
- INSERM U1103Université Clermont AuvergneCNRS UMR‐6293GReD InstituteTeam‐VolleClermont‐FerrandF‐63037France
| | - Hélène Holota
- INSERM U1103Université Clermont AuvergneCNRS UMR‐6293GReD InstituteTeam‐VolleClermont‐FerrandF‐63037France
| | - Mélusine Monrose
- INSERM U1103Université Clermont AuvergneCNRS UMR‐6293GReD InstituteTeam‐VolleClermont‐FerrandF‐63037France
| | - Manon Garcia
- INSERM U1103Université Clermont AuvergneCNRS UMR‐6293GReD InstituteTeam‐VolleClermont‐FerrandF‐63037France
| | - Angélique de Haze
- INSERM U1103Université Clermont AuvergneCNRS UMR‐6293GReD InstituteTeam‐VolleClermont‐FerrandF‐63037France
| | | | - Yoan Renaud
- INSERM U1103Université Clermont AuvergneCNRS UMR‐6293GReD InstituteBio‐informatic facilityClermont‐FerrandF‐63037France
| | - Jean‐Paul Saru
- INSERM U1103Université Clermont AuvergneCNRS UMR‐6293GReD InstituteTeam‐VolleClermont‐FerrandF‐63037France
| | - Alessia Perino
- Laboratory of Metabolic SignalingInstitute of BioengineeringSchool of Life SciencesEcole Polytechnique Fédérale de LausanneLausanneCH‐1015Switzerland
| | - Kristina Schoonjans
- Laboratory of Metabolic SignalingInstitute of BioengineeringSchool of Life SciencesEcole Polytechnique Fédérale de LausanneLausanneCH‐1015Switzerland
| | - Claude Beaudoin
- INSERM U1103Université Clermont AuvergneCNRS UMR‐6293GReD InstituteTeam‐VolleClermont‐FerrandF‐63037France
| | - David H. Volle
- INSERM U1103Université Clermont AuvergneCNRS UMR‐6293GReD InstituteTeam‐VolleClermont‐FerrandF‐63037France
| |
Collapse
|
10
|
Analysis of the Reversible Impact of the Chemodrug Busulfan on Mouse Testes. Cells 2021; 10:cells10092403. [PMID: 34572051 PMCID: PMC8472455 DOI: 10.3390/cells10092403] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 09/03/2021] [Accepted: 09/09/2021] [Indexed: 12/21/2022] Open
Abstract
Spermatogenesis is a process within the testis that leads to the production of spermatozoa. It is based on a population of spermatogonial stem cells, which have the capacity to self-renew and to differentiate throughout life to ensure the functions of reproduction are maintained. Male fertility disorders are responsible for half of the cases of infertility in couples worldwide. It is well known that cancer treatments are associated with reversible or irreversible fertility disorders. Busulfan (Bu) is an alkylating agent that significantly inhibits spermatogenesis. The present study relied on a combination of in vivo and in vitro approaches as well as RNAseq analysis to characterize the effects of Bu, in which mouse testes were used as a model. An in silico analysis revealed that many of the Bu-modulated genes are potentially regulated by the SIN3 Transcription Regulator Family Member A (SIN3A) and E2F Transcription Factor (E2F) families of transcription factors. The results demonstrate that the deregulated genes function in processes related to the cell cycle, DNA repair, and cell death mechanisms, including the Tumor Protein 53 (TP53) pathway. This reinforces the role of the TP53 signaling pathway as a major player in Bu effects. In addition, Bu altered the patterns of mRNA accumulation for various genes in undifferentiated spermatogonia. This work provides significant insight into the kinetics and impacts of busulfan, which could pave the way for developing strategies to minimize the impact of chemodrugs and, thus, could lead to germ cell lineage regeneration following anticancer treatments.
Collapse
|
11
|
Kothandapani A, Larsen MC, Lee J, Jorgensen JS, Jefcoate CR. Distinctive functioning of STARD1 in the fetal Leydig cells compared to adult Leydig and adrenal cells. Impact of Hedgehog signaling via the primary cilium. Mol Cell Endocrinol 2021; 531:111265. [PMID: 33864885 DOI: 10.1016/j.mce.2021.111265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 03/19/2021] [Accepted: 03/25/2021] [Indexed: 10/21/2022]
Abstract
STARD1 stimulates cholesterol transfer to mitochondrial CYP11A1 for conversion to pregnenolone. A cholesterol-binding START domain is guided by an N-terminal domain in a cell selective manner. Fetal and adult Leydig cells (FLC, ALC) show distinct Stard1 regulation. sm- FISH microscopy, which resolves individual molecules of Stard1 mRNA, shows uniformly high basal expression in each FLC. In ALC, in vivo, and cultured MA-10 cells, basal Stard1 expression is minimal. PKA activates loci asynchronously, with delayed splicing/export of 3.5 kb mRNA to mitochondria. After 60 min, ALC transition to an integrated mRNA delivery to mitochondria that is seen in FLC. Sertoli cells cooperate in Stard1 stimulation in FLC by delivering DHH to the primary cilium. There PTCH, SMO and cholesterol cooperate to release GLI3 to activate the Stard1 locus, probably by directing histone changes. ALC lack cilia. PKA then primes locus activation. FLC and ALC share similar SIK/CRTC/CREB regulation characterized for adrenal cells.
Collapse
Affiliation(s)
- Anbarasi Kothandapani
- Department of Comparative Biosciences, University of Wisconsin School of Veterinary Medicine, Madison, WI, 53706, USA
| | - Michele Campaigne Larsen
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Jinwoo Lee
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Joan S Jorgensen
- Department of Comparative Biosciences, University of Wisconsin School of Veterinary Medicine, Madison, WI, 53706, USA
| | - Colin R Jefcoate
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA.
| |
Collapse
|
12
|
Appanna N, Gibson H, Gangitano E, Dempster NJ, Morris K, George S, Arvaniti A, Gathercole LL, Keevil B, Penning TM, Storbeck KH, Tomlinson JW, Nikolaou N. Differential activity and expression of human 5β-reductase (AKR1D1) splice variants. J Mol Endocrinol 2021; 66:181-194. [PMID: 33502336 PMCID: PMC7965358 DOI: 10.1530/jme-20-0160] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 01/12/2021] [Indexed: 12/18/2022]
Abstract
Steroid hormones, including glucocorticoids and androgens, exert a wide variety of effects in the body across almost all tissues. The steroid A-ring 5β-reductase (AKR1D1) is expressed in human liver and testes, and three splice variants have been identified (AKR1D1-001, AKR1D1-002, AKR1D1-006). Amongst these, AKR1D1-002 is the best described; it modulates steroid hormone availability and catalyses an important step in bile acid biosynthesis. However, specific activity and expression of AKR1D1-001 and AKR1D1-006 are unknown. Expression of AKR1D1 variants were measured in human liver biopsies and hepatoma cell lines by qPCR. Their three-dimensional (3D) structures were predicted using in silico approaches. AKR1D1 variants were overexpressed in HEK293 cells, and successful overexpression confirmed by qPCR and Western blotting. Cells were treated with either cortisol, dexamethasone, prednisolone, testosterone or androstenedione, and steroid hormone clearance was measured by mass spectrometry. Glucocorticoid and androgen receptor activation were determined by luciferase reporter assays. AKR1D1-002 and AKR1D1-001 are expressed in human liver, and only AKR1D1-006 is expressed in human testes. Following overexpression, AKR1D1-001 and AKR1D1-006 protein levels were lower than AKR1D1-002, but significantly increased following treatment with the proteasomal inhibitor, MG-132. AKR1D1-002 efficiently metabolised glucocorticoids and androgens and decreased receptor activation. AKR1D1-001 and AKR1D1-006 poorly metabolised dexamethasone, but neither protein metabolised cortisol, prednisolone, testosterone or androstenedione. We have demonstrated the differential expression and role of AKR1D1 variants in steroid hormone clearance and receptor activation in vitro. AKR1D1-002 is the predominant functional protein in steroidogenic and metabolic tissues. In addition, AKR1D1-001 and AKR1D1-006 may have a limited, steroid-specific role in the regulation of dexamethasone action.
Collapse
Affiliation(s)
- Nathan Appanna
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford, Oxfordshire, UK
| | - Hylton Gibson
- Department of Biochemistry, Stellenbosch University, Stellenbosch, Western Cape, South Africa
| | - Elena Gangitano
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford, Oxfordshire, UK
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Lazio, Italy
| | - Niall J Dempster
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford, Oxfordshire, UK
| | - Karen Morris
- Biochemistry Department, Manchester University NHS Trust, Manchester Academic Health Science Centre, Manchester, Greater Manchester, UK
| | - Sherly George
- Biochemistry Department, Manchester University NHS Trust, Manchester Academic Health Science Centre, Manchester, Greater Manchester, UK
| | - Anastasia Arvaniti
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford, Oxfordshire, UK
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, Oxfordshire, UK
| | - Laura L Gathercole
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford, Oxfordshire, UK
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, Oxfordshire, UK
| | - Brian Keevil
- Biochemistry Department, Manchester University NHS Trust, Manchester Academic Health Science Centre, Manchester, Greater Manchester, UK
| | - Trevor M Penning
- Center of Excellence in Environmental Toxicology and Department of Systems Pharmacology & Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Karl-Heinz Storbeck
- Department of Biochemistry, Stellenbosch University, Stellenbosch, Western Cape, South Africa
| | - Jeremy W Tomlinson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford, Oxfordshire, UK
| | - Nikolaos Nikolaou
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford, Oxfordshire, UK
- Correspondence should be addressed to N Nikolaou:
| |
Collapse
|
13
|
Holota H, Thirouard L, Monrose M, Garcia M, De Haze A, Saru JP, Caira F, Beaudoin C, Volle DH. FXRα modulates leydig cell endocrine function in mouse. Mol Cell Endocrinol 2020; 518:110995. [PMID: 32827571 DOI: 10.1016/j.mce.2020.110995] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 07/27/2020] [Accepted: 08/14/2020] [Indexed: 01/14/2023]
Abstract
The hypothalamic-pituitary axis exert a major control over endocrine and exocrine testicular functions. The hypothalamic-pituitary axis corresponds to a cascade with the Gonadotropin Releasing Hormone secreted by the hypothalamus, which stimulates the synthesis and the release of Luteinizing Hormone (LH) and Follicle Stimulating Hormone by the gonadotropic cells of the anterior pituitary. The LH signaling pathway controls the steroidogenic activity of the Leydig cells via the activation of the luteinizing hormone/choriogonadotropin receptor. In order to avoid a runaway system, sex steroids exert a negative feedback within hypothalamus and pituitary. Testicular steroidogenesis is locally controlled within Leydig cells. The present work reviews some local regulations of steroidogenesis within the Leydig cells focusing mainly on the roles of the Farnesoid-X-Receptor-alpha and its interactions with several orphan members of the nuclear receptor superfamily. Further studies are required to reinforce our knowledge of the regulation of testicular endocrine function, which is necessary to ensure a better understanding of fertility disorders and then proposed an adequate treatment of the diseases.
Collapse
Affiliation(s)
- Hélène Holota
- Inserm U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD, F-63001, Clermont-Ferrand, France
| | - Laura Thirouard
- Inserm U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD, F-63001, Clermont-Ferrand, France
| | - Mélusine Monrose
- Inserm U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD, F-63001, Clermont-Ferrand, France
| | - Manon Garcia
- Inserm U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD, F-63001, Clermont-Ferrand, France
| | - Angélique De Haze
- Inserm U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD, F-63001, Clermont-Ferrand, France
| | - Jean-Paul Saru
- Inserm U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD, F-63001, Clermont-Ferrand, France
| | - Françoise Caira
- Inserm U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD, F-63001, Clermont-Ferrand, France
| | - Claude Beaudoin
- Inserm U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD, F-63001, Clermont-Ferrand, France
| | - David H Volle
- Inserm U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD, F-63001, Clermont-Ferrand, France.
| |
Collapse
|
14
|
Larsen MC, Lee J, Jorgensen JS, Jefcoate CR. STARD1 Functions in Mitochondrial Cholesterol Metabolism and Nascent HDL Formation. Gene Expression and Molecular mRNA Imaging Show Novel Splicing and a 1:1 Mitochondrial Association. Front Endocrinol (Lausanne) 2020; 11:559674. [PMID: 33193082 PMCID: PMC7607000 DOI: 10.3389/fendo.2020.559674] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022] Open
Abstract
STARD1 moves cholesterol (CHOL) from the outer mitochondrial membrane (OMM) to the inner membrane (IMM) in steroidogenic cells. This activity is integrated into CHOL trafficking and synthesis homeostasis, involving uptake through SR-B1 and LDL receptors and distribution through endosomes, ER, and lipid droplets. In adrenal cells, STARD1 is imported into the mitochondrial matrix accompanied by delivery of several hundred CHOL molecules. This transfer limits CYP11A1-mediated generation of pregnenolone. CHOL transfer is coupled to translation of STARD1 mRNA at the OMM. In testis cells, slower CHOL trafficking seems to be limiting. STARD1 also functions in a slower process through ER OMM contacts. The START domain of STARD1 is utilized by a family of genes, which includes additional STARD (forms 3-6) and GRAMD1B proteins that transfer CHOL. STARD forms 2 and 7 deliver phosphatidylcholine. STARD1 and STARD7 target their respective activities to mitochondria, via N-terminal domains (NTD) of over 50 amino acids. The NTD is not essential for steroidogenesis but exerts tissue-selective enhancement (testis>>adrenal). Three conserved sites for cleavage by the mitochondrial processing protease (MPP) generate three forms, each potentially with specific functions, as demonstrated in STARD7. STARD1 is expressed in macrophage and cardiac repair fibroblasts. Additional functions include CHOL metabolism by CYP27A1 that directs activation of LXR and CHOL export processes. STARD1 generates 3.5- and 1.6-kb mRNA from alternative polyadenylation. The 3.5-kb form exclusively binds the PKA-induced regulator, TIS11b, which binds at conserved sites in the extended 3'UTR to control mRNA translation and turnover. STARD1 expression also exhibits a novel, slow splicing that delayed splicing delivery of mRNA to mitochondria. Stimulation of transcription by PKA is directed by suppression of SIK forms that activate a CRTC/CREB/CBP promoter complex. This process is critical to pulsatile hormonal activation in vivo. sm-FISH RNA imaging shows a flow of single STARD1 mRNA particles from asymmetric accumulations of primary transcripts at gene loci to 1:1 complex of 3.5-kb mRNA with peri-nuclear mitochondria. Adrenal cells are similar but distinguished from testis cells by appreciable basal expression prior to hormonal activation. This difference is conserved in culture and in vivo.
Collapse
Affiliation(s)
- Michele Campaigne Larsen
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Jinwoo Lee
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
- Endocrinology and Reproductive Physiology Program, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Joan S. Jorgensen
- Endocrinology and Reproductive Physiology Program, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
- Department of Comparative Biosciences, University of Wisconsin School of Veterinary Medicine, Madison, WI, United States
| | - Colin R. Jefcoate
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
- Endocrinology and Reproductive Physiology Program, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| |
Collapse
|
15
|
Monrose M, Thirouard L, Garcia M, Holota H, De Haze A, Caira F, Beaudoin C, Volle DH. New perspectives on PPAR, VDR and FXRα as new actors in testicular pathophysiology. Mol Aspects Med 2020; 78:100886. [PMID: 32878696 DOI: 10.1016/j.mam.2020.100886] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/27/2020] [Accepted: 07/29/2020] [Indexed: 12/21/2022]
Abstract
The incidence of reproductive disorders is constantly increasing and affects 15% of couples, with male's abnormalities diagnosed in almost half of the cases. The male gonads exert two major functions of the testis with the productions of gametes (exocrine function) and of sexual hormones (endocrine function). In the last decades, next to steroid receptors such as estrogen and androgen receptors, the involvement of other members of the nuclear receptor superfamily have been described such as Steroidogenic factor-1 (SF-1), Nerve growth factor IB (NGFIB), Liver-X-Receptorα (LXRα) and Dosage-sensitive sex reversal, adrenal hypoplasia critical region, on chromosome X, gene 1 (DAX-1). The purpose of this review is to highlight the emerging roles of some members of the nuclear receptor superfamily among which the vitamin-D Receptor (VDR), Peroxisome Proliferator-Activated Receptor (PPAR), Farnesoid-X-Receptor-α (FXRα). We discuss how these receptors could participate to explain male fertility disorders; and their potential to be use as biomarkers or therapeutic targets for management of fertility disorders.
Collapse
Affiliation(s)
- M Monrose
- Inserm U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD, F-63001, Clermont-Ferrand, France
| | - L Thirouard
- Inserm U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD, F-63001, Clermont-Ferrand, France
| | - M Garcia
- Inserm U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD, F-63001, Clermont-Ferrand, France
| | - H Holota
- Inserm U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD, F-63001, Clermont-Ferrand, France
| | - A De Haze
- Inserm U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD, F-63001, Clermont-Ferrand, France
| | - F Caira
- Inserm U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD, F-63001, Clermont-Ferrand, France
| | - C Beaudoin
- Inserm U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD, F-63001, Clermont-Ferrand, France
| | - D H Volle
- Inserm U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD, F-63001, Clermont-Ferrand, France.
| |
Collapse
|
16
|
Agarwal D, Gireesh-Babu P, Pavan-Kumar A, Koringa P, Joshi CG, Chaudhari A. Transcriptome analysis of Clarias magur brain and gonads suggests neuro-endocrine inhibition of milt release from captive GnRH-induced males. Genomics 2020; 112:4041-4052. [PMID: 32650102 DOI: 10.1016/j.ygeno.2020.07.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/18/2020] [Accepted: 07/03/2020] [Indexed: 10/24/2022]
Abstract
Transcriptome analysis of Clarias magur brain and gonads at preparatory, mature, 6 and 16 h post-GnRH injection (hpi) stages yielded 9.5 GB data with 39,738 contigs. Sequences of 45 reproductive genes were identified for the first time in C. magur along with unique and differentially expressed genes. The expression of 20 genes was validated by qRT-PCR. Upregulation of Cyp11A1, Cyp17A1 and FTZF1 genes in the 16hpi testis accompanied by the 17β-HSD3 expression indicates testosterone (T) synthesis in response to LH surge, while reduced expression of CYP11B1 suggests a high T: 11-KT ratio. It is evident by the gene expression analysis that the inhibitory neurotransmitter GABA, altered T: 11-KT, increased testicular bile acids, and oxytocin-like neuropeptide in the male brain, appear to be involved in arresting the pulsatile motion of testicular smooth muscles. The work generates important leads for an effective induced breeding strategy for silurid catfish.
Collapse
Affiliation(s)
- Deepak Agarwal
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Fisheries Education, Mumbai, India
| | - P Gireesh-Babu
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Fisheries Education, Mumbai, India
| | - A Pavan-Kumar
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Fisheries Education, Mumbai, India
| | - Prakash Koringa
- Animal Biotechnology Department, Anand Agricultural University, Anand, India
| | - C G Joshi
- Animal Biotechnology Department, Anand Agricultural University, Anand, India
| | - Aparna Chaudhari
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Fisheries Education, Mumbai, India.
| |
Collapse
|
17
|
Garcia M, Thirouard L, Monrose M, Holota H, De Haze A, Caira F, Beaudoin C, Volle DH. Farnesoid X receptor alpha (FXRα) is a critical actor of the development and pathologies of the male reproductive system. Cell Mol Life Sci 2019; 76:4849-4859. [PMID: 31407019 PMCID: PMC11105758 DOI: 10.1007/s00018-019-03247-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/19/2019] [Accepted: 07/23/2019] [Indexed: 12/01/2022]
Abstract
The farnesoid-X-receptorα (FXRα; NR1H4) is one of the main bile acid (BA) receptors. During the last decades, through the use of pharmalogical approaches and transgenic mouse models, it has been demonstrated that the nuclear receptor FXRα controls numerous physiological functions such as glucose or energy metabolisms. It is also involved in the etiology or the development of several pathologies. Here, we will review the unexpected roles of FXRα on the male reproductive tract. FXRα has been demonstrated to play functions in the regulation of testicular and prostate homeostasis. Even though additional studies are needed to confirm these findings in humans, the reviewed reports open new field of research to better define the effects of bile acid-FXRα signaling pathways on fertility disorders and cancers.
Collapse
Affiliation(s)
- Manon Garcia
- Inserm U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD, 28 Place Henri Dunant, 63001, Clermont-Ferrand, France
| | - Laura Thirouard
- Inserm U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD, 28 Place Henri Dunant, 63001, Clermont-Ferrand, France
| | - Mélusine Monrose
- Inserm U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD, 28 Place Henri Dunant, 63001, Clermont-Ferrand, France
| | - Hélène Holota
- Inserm U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD, 28 Place Henri Dunant, 63001, Clermont-Ferrand, France
| | - Angélique De Haze
- Inserm U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD, 28 Place Henri Dunant, 63001, Clermont-Ferrand, France
| | - Françoise Caira
- Inserm U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD, 28 Place Henri Dunant, 63001, Clermont-Ferrand, France
| | - Claude Beaudoin
- Inserm U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD, 28 Place Henri Dunant, 63001, Clermont-Ferrand, France.
| | - David H Volle
- Inserm U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD, 28 Place Henri Dunant, 63001, Clermont-Ferrand, France.
| |
Collapse
|
18
|
Weger BD, Gobet C, Yeung J, Martin E, Jimenez S, Betrisey B, Foata F, Berger B, Balvay A, Foussier A, Charpagne A, Boizet-Bonhoure B, Chou CJ, Naef F, Gachon F. The Mouse Microbiome Is Required for Sex-Specific Diurnal Rhythms of Gene Expression and Metabolism. Cell Metab 2019; 29:362-382.e8. [PMID: 30344015 PMCID: PMC6370974 DOI: 10.1016/j.cmet.2018.09.023] [Citation(s) in RCA: 199] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 06/27/2018] [Accepted: 09/25/2018] [Indexed: 02/08/2023]
Abstract
The circadian clock and associated feeding rhythms have a profound impact on metabolism and the gut microbiome. To what extent microbiota reciprocally affect daily rhythms of physiology in the host remains elusive. Here, we analyzed transcriptome and metabolome profiles of male and female germ-free mice. While mRNA expression of circadian clock genes revealed subtle changes in liver, intestine, and white adipose tissue, germ-free mice showed considerably altered expression of genes associated with rhythmic physiology. Strikingly, the absence of the microbiome attenuated liver sexual dimorphism and sex-specific rhythmicity. The resulting feminization of male and masculinization of female germ-free animals is likely caused by altered sexual development and growth hormone secretion, associated with differential activation of xenobiotic receptors. This defines a novel mechanism by which the microbiome regulates host metabolism.
Collapse
Affiliation(s)
- Benjamin D Weger
- Department of Diabetes and Circadian Rhythms, Nestlé Institute of Health Sciences, 1015 Lausanne, Switzerland
| | - Cédric Gobet
- Department of Diabetes and Circadian Rhythms, Nestlé Institute of Health Sciences, 1015 Lausanne, Switzerland; Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Jake Yeung
- Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Eva Martin
- Department of Diabetes and Circadian Rhythms, Nestlé Institute of Health Sciences, 1015 Lausanne, Switzerland
| | - Sonia Jimenez
- Department of Diabetes and Circadian Rhythms, Nestlé Institute of Health Sciences, 1015 Lausanne, Switzerland
| | - Bertrand Betrisey
- Cellular Metabolism, Department of Cell Biology, Nestlé Institute of Health Sciences, Nestlé Research, 1015 Lausanne, Switzerland
| | - Francis Foata
- Host-Microbe Interaction, Department of Gastro-Intestinal Health, Nestlé Institute of Health Sciences, Nestlé Research, 1000 Lausanne, Switzerland
| | - Bernard Berger
- Host-Microbe Interaction, Department of Gastro-Intestinal Health, Nestlé Institute of Health Sciences, Nestlé Research, 1000 Lausanne, Switzerland
| | - Aurélie Balvay
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Anne Foussier
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Aline Charpagne
- Genomics, Department of Multi-Omics, Nestlé Institute of Health Sciences, Nestlé Research, 1015 Lausanne, Switzerland
| | - Brigitte Boizet-Bonhoure
- Institut de Génétique Humaine, CNRS-Université de Montpellier UMR9002, 34396 Montpellier, France
| | - Chieh Jason Chou
- Host-Microbe Interaction, Department of Gastro-Intestinal Health, Nestlé Institute of Health Sciences, Nestlé Research, 1000 Lausanne, Switzerland
| | - Felix Naef
- Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Frédéric Gachon
- Department of Diabetes and Circadian Rhythms, Nestlé Institute of Health Sciences, 1015 Lausanne, Switzerland; School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland.
| |
Collapse
|
19
|
Nuclear Receptor Metabolism of Bile Acids and Xenobiotics: A Coordinated Detoxification System with Impact on Health and Diseases. Int J Mol Sci 2018; 19:ijms19113630. [PMID: 30453651 PMCID: PMC6274770 DOI: 10.3390/ijms19113630] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 11/14/2018] [Accepted: 11/14/2018] [Indexed: 02/06/2023] Open
Abstract
Structural and functional studies have provided numerous insights over the past years on how members of the nuclear hormone receptor superfamily tightly regulate the expression of drug-metabolizing enzymes and transporters. Besides the role of the farnesoid X receptor (FXR) in the transcriptional control of bile acid transport and metabolism, this review provides an overview on how this metabolic sensor prevents the accumulation of toxic byproducts derived from endogenous metabolites, as well as of exogenous chemicals, in coordination with the pregnane X receptor (PXR) and the constitutive androstane receptor (CAR). Decrypting this network should provide cues to better understand how these metabolic nuclear receptors participate in physiologic and pathologic processes with potential validation as therapeutic targets in human disabilities and cancers.
Collapse
|
20
|
Sèdes L, Desdoits-Lethimonier C, Rouaisnel B, Holota H, Thirouard L, Lesne L, Damon-Soubeyrand C, Martinot E, Saru JP, Mazaud-Guittot S, Caira F, Beaudoin C, Jégou B, Volle DH. Crosstalk between BPA and FXRα Signaling Pathways Lead to Alterations of Undifferentiated Germ Cell Homeostasis and Male Fertility Disorders. Stem Cell Reports 2018; 11:944-958. [PMID: 30245210 PMCID: PMC6178796 DOI: 10.1016/j.stemcr.2018.08.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 08/27/2018] [Accepted: 08/27/2018] [Indexed: 12/20/2022] Open
Abstract
Several studies have reported an association between the farnesoid X receptor alpha (FXRα) and estrogenic signaling pathways. Fxrα could thus be involved in the reprotoxic effects of endocrine disruptors such as bisphenol-A (BPA). To test this hypothesis, mice were exposed to BPA and/or stigmasterol (S), an FXRα antagonist. Following the exposure to both molecules, wild-type animals showed impaired fertility and lower sperm cell production associated with the alteration of the establishment and maintenance of the undifferentiated germ cell pool. The crosstalk between BPA and FXRα is further supported by the lower impact of BPA in mice genetically ablated for Fxrα and the fact that BPA counteracted the effects of FXRα agonists. These effects might result from the downregulation of Fxrα expression following BPA exposure. BPA and S act additively in human testis. Our data demonstrate that FXRα activity modulates the impact of BPA on male gonads and on undifferentiated germ cell population. BPA and S exposures synergistically induce male fertility disorders BPA regulates Fxr expression BPA and S act additively in human testis
Collapse
Affiliation(s)
- Lauriane Sèdes
- INSERM U 1103, Université Clermont Auvergne, CNRS, UMR 6293, GReD, Laboratoire Génétique, Reproduction & Développement, 28 Place Henri-Dunant, 63000 Clermont-Ferrand, France
| | - Christèle Desdoits-Lethimonier
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Betty Rouaisnel
- INSERM U 1103, Université Clermont Auvergne, CNRS, UMR 6293, GReD, Laboratoire Génétique, Reproduction & Développement, 28 Place Henri-Dunant, 63000 Clermont-Ferrand, France
| | - Hélène Holota
- INSERM U 1103, Université Clermont Auvergne, CNRS, UMR 6293, GReD, Laboratoire Génétique, Reproduction & Développement, 28 Place Henri-Dunant, 63000 Clermont-Ferrand, France
| | - Laura Thirouard
- INSERM U 1103, Université Clermont Auvergne, CNRS, UMR 6293, GReD, Laboratoire Génétique, Reproduction & Développement, 28 Place Henri-Dunant, 63000 Clermont-Ferrand, France
| | - Laurianne Lesne
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Christelle Damon-Soubeyrand
- INSERM U 1103, Université Clermont Auvergne, CNRS, UMR 6293, GReD, Laboratoire Génétique, Reproduction & Développement, 28 Place Henri-Dunant, 63000 Clermont-Ferrand, France
| | - Emmanuelle Martinot
- INSERM U 1103, Université Clermont Auvergne, CNRS, UMR 6293, GReD, Laboratoire Génétique, Reproduction & Développement, 28 Place Henri-Dunant, 63000 Clermont-Ferrand, France
| | - Jean-Paul Saru
- INSERM U 1103, Université Clermont Auvergne, CNRS, UMR 6293, GReD, Laboratoire Génétique, Reproduction & Développement, 28 Place Henri-Dunant, 63000 Clermont-Ferrand, France
| | - Séverine Mazaud-Guittot
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Françoise Caira
- INSERM U 1103, Université Clermont Auvergne, CNRS, UMR 6293, GReD, Laboratoire Génétique, Reproduction & Développement, 28 Place Henri-Dunant, 63000 Clermont-Ferrand, France
| | - Claude Beaudoin
- INSERM U 1103, Université Clermont Auvergne, CNRS, UMR 6293, GReD, Laboratoire Génétique, Reproduction & Développement, 28 Place Henri-Dunant, 63000 Clermont-Ferrand, France
| | - Bernard Jégou
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - David H Volle
- INSERM U 1103, Université Clermont Auvergne, CNRS, UMR 6293, GReD, Laboratoire Génétique, Reproduction & Développement, 28 Place Henri-Dunant, 63000 Clermont-Ferrand, France.
| |
Collapse
|
21
|
Yang T, Mei H, Xu D, Zhou W, Zhu X, Sun L, Huang X, Wang X, Shu T, Liu J, Ding J, Hassan HM, Zhang L, Jiang Z. Early indications of ANIT-induced cholestatic liver injury: Alteration of hepatocyte polarization and bile acid homeostasis. Food Chem Toxicol 2017; 110:1-12. [PMID: 28986171 DOI: 10.1016/j.fct.2017.09.051] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 09/04/2017] [Accepted: 09/29/2017] [Indexed: 02/08/2023]
Abstract
Hepatocyte polarization is essential for biliary secretion, and loss of polarity causes bile secretory failure and hepatotoxicity. Here, we showed that alpha-naphthyl isothiocyanate (ANIT)-induced liver injury was accompanied by the dynamic interruption of bile acid homeostasis in rat plasma, liver and bile, which was characterized by the redistribution of bile acids in plasma and bile and a small range of fluctuations in the liver. Molecular mechanism studies indicated that these factors are dynamically mediated by the disruption of bile acid transporters and hepatic tight junctions. Dynamic changes in tight junction (TJ) permeability were observed by hepatobiliary barrier function assessment. Hepatocyte polarization was disrupted by ANIT before the development of cholestatic hepatotoxicity and alteration of bile acid metabolic profiles, which were assayed by high-performance liquid chromatography-tandem mass spectrometry, further verifying TJ deficiency. S1PR1 activation with SEW2871 reduced ANIT-induced liver injury by reducing the total serum bile acid concentration, liver functional enzyme activity and inflammation. Our data suggest that hepatocyte polarization plays an important role in maintaining bile acid homeostasis before the development of cholestatic hepatotoxicity and that TJs were more prominent in the early stage of cholestasis. S1PR1 may be a potential target for the prevention of drug-induced cholestatic liver injury.
Collapse
Affiliation(s)
- Tingting Yang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Huifang Mei
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Dengqiu Xu
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Wang Zhou
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaoyu Zhu
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Lixin Sun
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Xin Huang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Xue Wang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Ting Shu
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Jia Liu
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Jiaxin Ding
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - H M Hassan
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Luyong Zhang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Center for Drug Screening and Pharmacodynamics Evaluation, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Zhenzhou Jiang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Ministry of Education, Nanjing 210009, China.
| |
Collapse
|