1
|
Liu X, Chen X, Wang C, Song J, Xu J, Gao Z, Huang Y, Suo H. Mechanisms of probiotic modulation of ovarian sex hormone production and metabolism: a review. Food Funct 2024; 15:2860-2878. [PMID: 38433710 DOI: 10.1039/d3fo04345b] [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: 03/05/2024]
Abstract
Sex hormones play a pivotal role in the growth and development of the skeletal, neurological, and reproductive systems. In women, the dysregulation of sex hormones can result in various health complications such as acne, hirsutism, and irregular menstruation. One of the most prevalent diseases associated with excess androgens is polycystic ovary syndrome with a hyperandrogenic phenotype. Probiotics have shown the potential to enhance the secretion of ovarian sex hormones. However, the underlying mechanism of action remains unclear. Furthermore, comprehensive reviews detailing how probiotics modulate ovarian sex hormones are scarce. This review seeks to shed light on the potential mechanisms through which probiotics influence the production of ovarian sex hormones. The role of probiotics across various biological axes, including the gut-ovarian, gut-brain-ovarian, gut-liver-ovarian, gut-pancreas-ovarian, and gut-fat-ovarian axes, with a focus on the direct impact of probiotics on the ovaries via the gut and their effects on brain gonadotropins is discussed. It is also proposed herein that probiotics can significantly influence the onset, progression, and complications of ovarian sex hormone abnormalities. In addition, this review provides a theoretical basis for the therapeutic application of probiotics in managing sex hormone-related health conditions.
Collapse
Affiliation(s)
- Xiao Liu
- College of Food Science, Southwest University, Chongqing 400715, P. R. China.
| | - Xiaoyong Chen
- College of Food Science, Southwest University, Chongqing 400715, P. R. China.
- Citrus Research Institute, National Citrus Engineering Research Center, Southwest University, Chongqing 400715, P. R. China
- Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, P. R. China
| | - Chen Wang
- College of Food Science, Southwest University, Chongqing 400715, P. R. China.
- Citrus Research Institute, National Citrus Engineering Research Center, Southwest University, Chongqing 400715, P. R. China
- Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, P. R. China
| | - Jiajia Song
- College of Food Science, Southwest University, Chongqing 400715, P. R. China.
- Citrus Research Institute, National Citrus Engineering Research Center, Southwest University, Chongqing 400715, P. R. China
- Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, P. R. China
| | - Jiahui Xu
- College of Food Science, Southwest University, Chongqing 400715, P. R. China.
| | - Zhen Gao
- College of Food Science, Southwest University, Chongqing 400715, P. R. China.
| | - Yechuan Huang
- College of Bioengineering, Jingchu University of Technology, Jingmen 448000, P. R. China.
| | - Huayi Suo
- College of Food Science, Southwest University, Chongqing 400715, P. R. China.
- Citrus Research Institute, National Citrus Engineering Research Center, Southwest University, Chongqing 400715, P. R. China
- Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, P. R. China
| |
Collapse
|
2
|
Rusch JA, Layden BT, Dugas LR. Signalling cognition: the gut microbiota and hypothalamic-pituitary-adrenal axis. Front Endocrinol (Lausanne) 2023; 14:1130689. [PMID: 37404311 PMCID: PMC10316519 DOI: 10.3389/fendo.2023.1130689] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 05/25/2023] [Indexed: 07/06/2023] Open
Abstract
Cognitive function in humans depends on the complex and interplay between multiple body systems, including the hypothalamic-pituitary-adrenal (HPA) axis. The gut microbiota, which vastly outnumbers human cells and has a genetic potential that exceeds that of the human genome, plays a crucial role in this interplay. The microbiota-gut-brain (MGB) axis is a bidirectional signalling pathway that operates through neural, endocrine, immune, and metabolic pathways. One of the major neuroendocrine systems responding to stress is the HPA axis which produces glucocorticoids such as cortisol in humans and corticosterone in rodents. Appropriate concentrations of cortisol are essential for normal neurodevelopment and function, as well as cognitive processes such as learning and memory, and studies have shown that microbes modulate the HPA axis throughout life. Stress can significantly impact the MGB axis via the HPA axis and other pathways. Animal research has advanced our understanding of these mechanisms and pathways, leading to a paradigm shift in conceptual thinking about the influence of the microbiota on human health and disease. Preclinical and human trials are currently underway to determine how these animal models translate to humans. In this review article, we summarize the current knowledge of the relationship between the gut microbiota, HPA axis, and cognition, and provide an overview of the main findings and conclusions in this broad field.
Collapse
Affiliation(s)
- Jody A. Rusch
- Division of Chemical Pathology, Department of Pathology, University of Cape Town, Cape Town, South Africa
- C17 Chemical Pathology Laboratory, Groote Schuur Hospital, National Health Laboratory Service, Cape Town, South Africa
| | - Brian T. Layden
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
- Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States
| | - Lara R. Dugas
- Division of Epidemiology and Biostatistics, School of Public Health, University of Cape Town, Cape Town, South Africa
- Public Health Sciences, Parkinson School of Health Sciences and Public Health, Loyola University Chicago, Maywood, IL, United States
| |
Collapse
|
3
|
Ahmed M, Mäkinen VP, Lumsden A, Boyle T, Mulugeta A, Lee SH, Olver I, Hyppönen E. Metabolic profile predicts incident cancer: A large-scale population study in the UK Biobank. Metabolism 2023; 138:155342. [PMID: 36377121 DOI: 10.1016/j.metabol.2022.155342] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/24/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND AND AIMS Analyses to predict the risk of cancer typically focus on single biomarkers, which do not capture their complex interrelations. We hypothesized that the use of metabolic profiles may provide new insights into cancer prediction. METHODS We used information from 290,888 UK Biobank participants aged 37 to 73 years at baseline. Metabolic subgroups were defined based on clustering of biochemical data using an artificial neural network approach and examined for their association with incident cancers identified through linkage to cancer registry. In addition, we evaluated associations between 38 individual biomarkers and cancer risk. RESULTS In total, 21,973 individuals developed cancer during the follow-up (median 3.87 years, interquartile range [IQR] = 2.03-5.58). Compared to the metabolically favorable subgroup (IV), subgroup III (defined as "high BMI, C-reactive protein & cystatin C") was associated with a higher risk of obesity-related cancers (hazard ratio [HR] = 1.26, 95 % CI = 1.21 to 1.32) and hematologic-malignancies (e.g., lymphoid leukemia: HR = 1.83, 95%CI = 1.44 to 2.33). Subgroup II ("high triglycerides & liver enzymes") was strongly associated with liver cancer risk (HR = 5.70, 95%CI = 3.57 to 9.11). Analysis of individual biomarkers showed a positive association between testosterone and greater risks of hormone-sensitive cancers (HR per SD higher = 1.32, 95%CI = 1.23 to 1.44), and liver cancer (HR = 2.49, 95%CI =1.47 to 4.24). Many liver tests were individually associated with a greater risk of liver cancer with the strongest association observed for gamma-glutamyl transferase (HR = 2.40, 95%CI = 2.19 to 2.65). CONCLUSIONS Metabolic profile in middle-to-older age can predict cancer incidence, in particular risk of obesity-related cancer, hematologic malignancies, and liver cancer. Elevated values from liver tests are strong predictors for later risk of liver cancer.
Collapse
Affiliation(s)
- Muktar Ahmed
- Australian Centre for Precision Health, University of South Australia, Adelaide, SA, Australia; Department of Epidemiology, Faculty of Public Health, Jimma University Institute of Health, Jimma, Ethiopia; UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia; South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Ville-Petteri Mäkinen
- Australian Centre for Precision Health, University of South Australia, Adelaide, SA, Australia; Computational Systems Biology Program, Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Amanda Lumsden
- Australian Centre for Precision Health, University of South Australia, Adelaide, SA, Australia; UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Terry Boyle
- Australian Centre for Precision Health, University of South Australia, Adelaide, SA, Australia; South Australian Health and Medical Research Institute, Adelaide, SA, Australia; UniSA Allied Health & Human Performance, University of South Australia, Adelaide, SA, Australia
| | - Anwar Mulugeta
- Australian Centre for Precision Health, University of South Australia, Adelaide, SA, Australia; UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia; South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Sang Hong Lee
- Australian Centre for Precision Health, University of South Australia, Adelaide, SA, Australia; South Australian Health and Medical Research Institute, Adelaide, SA, Australia; UniSA Allied Health & Human Performance, University of South Australia, Adelaide, SA, Australia
| | - Ian Olver
- School of Psychology, Faculty of Health and Medical Sciences, University of Adelaide, Australia
| | - Elina Hyppönen
- Australian Centre for Precision Health, University of South Australia, Adelaide, SA, Australia; UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia; South Australian Health and Medical Research Institute, Adelaide, SA, Australia.
| |
Collapse
|
4
|
Panzitt K, Zollner G, Marschall HU, Wagner M. Recent advances on FXR-targeting therapeutics. Mol Cell Endocrinol 2022; 552:111678. [PMID: 35605722 DOI: 10.1016/j.mce.2022.111678] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 12/25/2022]
Abstract
The bile acid receptor FXR has emerged as a bona fide drug target for chronic cholestatic and metabolic liver diseases, ahead of all non-alcoholic fatty liver disease (NAFLD). FXR is highly expressed in the liver and intestine and activation at both sites differentially contributes to its desired metabolic effects. Unrestricted FXR activation, however, also comes along with undesired effects such as a pro-atherogenic lipid profile, pruritus and hepatocellular toxicity under certain conditions. Several pre-clinical studies have confirmed the potency of FXR activation for cholestatic and metabolic liver diseases, but overall it remains still open whether selective activation of intestinal FXR is advantageous over pan-FXR activation and whether restricted or modulated FXR activation can limit some of the side effects. Even more, FXR antagonist also bear the potential as intestinal-selective drugs in NAFLD models. In this review we will discuss the molecular prerequisites for FXR activation, pan-FXR activation and intestinal FXR in/activation from a therapeutic point of view, different steroidal and non-steroidal FXR agonists, ways to restrict FXR activation and finally what we have learned from pre-clinical models and clinical trials with different FXR therapeutics.
Collapse
Affiliation(s)
- Katrin Panzitt
- Research Unit for Translational Nuclear Receptor Research, Medical University Graz, Graz, Austria; Division of Gastroenterology and Hepatology, Medical University Graz, Graz, Austria
| | - Gernot Zollner
- Division of Gastroenterology and Hepatology, Medical University Graz, Graz, Austria
| | - Hanns-Ulrich Marschall
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Martin Wagner
- Research Unit for Translational Nuclear Receptor Research, Medical University Graz, Graz, Austria; Division of Gastroenterology and Hepatology, Medical University Graz, Graz, Austria.
| |
Collapse
|
5
|
FXR in liver physiology: Multiple faces to regulate liver metabolism. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166133. [PMID: 33771667 DOI: 10.1016/j.bbadis.2021.166133] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 03/11/2021] [Accepted: 03/17/2021] [Indexed: 12/15/2022]
Abstract
The liver is the central metabolic hub which coordinates nutritional inputs and metabolic outputs. Food intake releases bile acids which can be sensed by the bile acid receptor FXR in the liver and the intestine. Hepatic and intestinal FXR coordinately regulate postprandial nutrient disposal in a network of interacting metabolic nuclear receptors. In this review we summarize and update the "classical roles" of FXR as a central integrator of the feeding state response, which orchestrates the metabolic processing of carbohydrates, lipids, proteins and bile acids. We also discuss more recent and less well studied FXR effects on amino acid, protein metabolism, autophagic turnover and inflammation. In addition, we summarize the recent understanding of how FXR signaling is affected by posttranslational modifications and by different FXR isoforms. These modifications and variations in FXR signaling might be considered when FXR is targeted pharmaceutically in clinical applications.
Collapse
|
6
|
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: 2.5] [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
|
7
|
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
|
8
|
Holota H, Thirouard L, Garcia M, Monrose M, de Haze A, Saru JP, Caira F, Beaudoin C, Volle DH. Fxralpha gene is a target gene of hCG signaling pathway and represses hCG induced steroidogenesis. J Steroid Biochem Mol Biol 2019; 194:105460. [PMID: 31470110 DOI: 10.1016/j.jsbmb.2019.105460] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 07/19/2019] [Accepted: 08/27/2019] [Indexed: 12/14/2022]
Abstract
The bile acid receptor Farnesoid-X-Receptor alpha (FXRα), a member of the nuclear receptor superfamily, is well known for its roles in the enterohepatic tract. In addition, FXRα regulates testicular physiology through the control of both endocrine and exocrine functions. The endocrine function of the Leydig cells is mainly controlled by the hypothalamo-pituitary axis viaLH/chorionic gonadotropin (CG). If FXRα was demonstrated to control the expression of the Lhcgr gene, encoding the LH receptor; the impact of the LH/CG signaling on the Fxrα expression has not been defined so far. Here, we demonstrate that hCG increases the Fxrα gene expression through the protein kinase-A signaling pathway. Fxrα is then involved in a negative feedback of steroid synthesis. These data improve our knowledge of the local control of the testicular steroidogenesis with the identification of the link between the hypothalamo-pituitary axis and the FXRα signaling pathway.
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
| | - Manon Garcia
- 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
| | - 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
|
9
|
Xiong XL, Ding Y, Chen ZL, Wang Y, Liu P, Qin H, Zhou LS, Zhang LL, Huang J, Zhao L. Emodin Rescues Intrahepatic Cholestasis via Stimulating FXR/BSEP Pathway in Promoting the Canalicular Export of Accumulated Bile. Front Pharmacol 2019; 10:522. [PMID: 31191298 PMCID: PMC6540617 DOI: 10.3389/fphar.2019.00522] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 04/25/2019] [Indexed: 12/14/2022] Open
Abstract
Aim Bile salt export pump (BSEP) have been confirmed to play an important role for bile acid canalicular export in the treatment of cholestasis. In this study, we investigated the stimulatory effect of emodin on BSEP signaling pathway in cholestasis. Methods Cell and animal experiments were given different concentrations of emodin. The BSEP upstream molecule farnesoid X receptor was down-regulated by small interfering RNA (siRNA) technology or guggulsterones and up-regulated by lentivirus or GW4064. Real-time PCR and Western blotting was employed to detect the mRNA and protein levels of BSEP in LO2 cell, rat primary hepatocytes and liver tissue. Immunohistochemistry (IHC) was used to examine the expression of BSEP in liver tissues. Rat liver function and pathological changes of liver tissue were performed by biochemical test and hematoxylin and eosin (HE) staining. Results Emodin could increase the mRNA and protein expression of BSEP and FXR. When down-regulating farnesoid X receptor expression with the siRNA or inhibitor guggulsterones, and up-regulating farnesoid X receptor expression with the lentivirus or agonist GW4064, emodin could increase the mRNA level of BSEP and FXR and the protein level of BSEP, FXR1, and FXR2. Emodin also had a notable effect on rat primary hepatocytes experiment, rat pathological manifestation, BSEP, FXR1, and FXR2 positive staining in liver tissues and the test of liver function. Conclusion Emodin has a protective effect and a rescue activity on cholestasis via stimulating FXR/BSEP pathways in promoting the canalicular export of accumulated bile.
Collapse
Affiliation(s)
- Xiao-Li Xiong
- Department of Integrated Chinese and Western Medicine, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Ding
- Department of Infectious Diseases and Immunology, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhi-Lin Chen
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yao Wang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Pan Liu
- School of First Clinical Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Huan Qin
- Department of Clinical Laboratory, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li-Shan Zhou
- Department of Integrated Chinese and Western Medicine, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ling-Ling Zhang
- Department of Integrated Chinese and Western Medicine, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Juan Huang
- Department of Pathology, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Zhao
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
10
|
Theiler-Schwetz V, Zaufel A, Schlager H, Obermayer-Pietsch B, Fickert P, Zollner G. Bile acids and glucocorticoid metabolism in health and disease. Biochim Biophys Acta Mol Basis Dis 2019; 1865:243-251. [DOI: 10.1016/j.bbadis.2018.08.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/18/2018] [Accepted: 08/01/2018] [Indexed: 12/12/2022]
|
11
|
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: 2.2] [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
|
12
|
Alwashih MA, Watson DG, Andrew R, Stimson RH, Alossaimi M, Blackburn G, Walker BR. Plasma metabolomic profile varies with glucocorticoid dose in patients with congenital adrenal hyperplasia. Sci Rep 2017; 7:17092. [PMID: 29213133 PMCID: PMC5719028 DOI: 10.1038/s41598-017-17220-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 11/23/2017] [Indexed: 11/09/2022] Open
Abstract
Glucocorticoid replacement therapy is the mainstay of treatment for congenital adrenal hyperplasia (CAH) but has a narrow therapeutic index and dose optimisation is challenging. Metabolomic profiling was carried out on plasma samples from 117 adults with 21-hydroxylase deficiency receiving their usual glucocorticoid replacement therapy who were part of the CaHASE study. Samples were profiled by using hydrophilic interaction chromatography with high resolution mass spectrometry. The patients were also profiled using nine routine clinical measures. The data were modelled by using both multivariate and univariate statistics by using the clinical metadata to inform the choice of patient groupings. Comparison of 382 metabolites amongst groups receiving different glucocorticoid doses revealed a clear distinction between patients receiving ≤5 mg (n = 64) and >5 mg (n = 53) daily prednisolone-equivalent doses. The 24 metabolites which were statistically significantly different between groups included free fatty acids, bile acids, and amino acid metabolites. Using 7 metabolites improved the receiver operating characteristic with area under the curve for predicting glucocorticoid dose of >0.9 with FDR adjusted P values in the range 3.3 E-04 -1.9 E-10. A combination of seven plasma metabolite biomarkers readily discriminates supraphysiological glucocorticoid replacement doses in patients with CAH.
Collapse
Affiliation(s)
- Mohammad A Alwashih
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE, UK.,General Directorate of Medical Services, Ministry of Interior, Riyadh, 13321, Saudi Arabia
| | - David G Watson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE, UK.
| | - Ruth Andrew
- BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Roland H Stimson
- BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Manal Alossaimi
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE, UK.,Ministry of Health, Riyadh, Saudi Arabia
| | - Gavin Blackburn
- Glasgow Polyomics, Wolfson Wohl Cancer Research Centre, College of Medical, Veterinary & Life Sciences, University of Glasgow, Garscube Estate Switchback Road, Bearsden, G61 1QH, UK
| | - Brian R Walker
- BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| |
Collapse
|
13
|
Baptissart M, Martinot E, Vega A, Sédes L, Rouaisnel B, de Haze A, Baron S, Schoonjans K, Caira F, Volle DH. Bile acid-FXRα pathways regulate male sexual maturation in mice. Oncotarget 2017; 7:19468-82. [PMID: 26848619 PMCID: PMC4991395 DOI: 10.18632/oncotarget.7153] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 01/22/2016] [Indexed: 12/21/2022] Open
Abstract
The bile acid receptor Farnesol-X-Receptor alpha (FRXα) is a member of the nuclear receptor superfamily. FRXα is expressed in the interstitial compartment of the adult testes, which contain the Leydig cells. In adult, short term treatment (12 hours) with FRXα agonist inhibits the expression of steroidogenic genes via the induction of the Small heterodimer partner (SHP). However the consequences of FRXα activation on testicular pathophysiology have never been evaluated. We demonstrate here that mice fed a diet supplemented with bile acid during pubertal age show increased incidence of infertility. This is associated with altered differentiation and increase apoptosis of germ cells due to lower testosterone levels. At the molecular level, next to the repression of basal steroidogenesis via the induction expression of Shp and Dax-1, two repressors of steroidogenesis, the main action of the BA-FRXα signaling is through lowering the Leydig cell sensitivity to the hypothalamo-pituitary axis, the main regulator of testicular endocrine function. In conclusion, BA-FRXα signaling is a critical actor during sexual maturation.
Collapse
Affiliation(s)
- Marine Baptissart
- INSERM U 1103, Laboratoire GReD, Campus Universitaire des Cézeaux, TSA 60026, CS 60026, 63178 Aubière Cedex, France.,Université Clermont Auvergne, Université Blaise Pascal, GReD, F-63178 Aubière, France.,CNRS, UMR 6293, GReD, F-63178 Aubière, France.,Centre de Recherche en Nutrition Humaine d'Auvergne, F-63000 Clermont-Ferrand, France
| | - Emmanuelle Martinot
- INSERM U 1103, Laboratoire GReD, Campus Universitaire des Cézeaux, TSA 60026, CS 60026, 63178 Aubière Cedex, France.,Université Clermont Auvergne, Université Blaise Pascal, GReD, F-63178 Aubière, France.,CNRS, UMR 6293, GReD, F-63178 Aubière, France.,Centre de Recherche en Nutrition Humaine d'Auvergne, F-63000 Clermont-Ferrand, France
| | - Aurélie Vega
- INSERM U 1103, Laboratoire GReD, Campus Universitaire des Cézeaux, TSA 60026, CS 60026, 63178 Aubière Cedex, France.,Université Clermont Auvergne, Université Blaise Pascal, GReD, F-63178 Aubière, France.,CNRS, UMR 6293, GReD, F-63178 Aubière, France.,Centre de Recherche en Nutrition Humaine d'Auvergne, F-63000 Clermont-Ferrand, France
| | - Lauriane Sédes
- INSERM U 1103, Laboratoire GReD, Campus Universitaire des Cézeaux, TSA 60026, CS 60026, 63178 Aubière Cedex, France.,Université Clermont Auvergne, Université Blaise Pascal, GReD, F-63178 Aubière, France.,CNRS, UMR 6293, GReD, F-63178 Aubière, France.,Centre de Recherche en Nutrition Humaine d'Auvergne, F-63000 Clermont-Ferrand, France
| | - Betty Rouaisnel
- INSERM U 1103, Laboratoire GReD, Campus Universitaire des Cézeaux, TSA 60026, CS 60026, 63178 Aubière Cedex, France.,Université Clermont Auvergne, Université Blaise Pascal, GReD, F-63178 Aubière, France.,CNRS, UMR 6293, GReD, F-63178 Aubière, France.,Centre de Recherche en Nutrition Humaine d'Auvergne, F-63000 Clermont-Ferrand, France
| | - Angélique de Haze
- INSERM U 1103, Laboratoire GReD, Campus Universitaire des Cézeaux, TSA 60026, CS 60026, 63178 Aubière Cedex, France.,Université Clermont Auvergne, Université Blaise Pascal, GReD, F-63178 Aubière, France.,CNRS, UMR 6293, GReD, F-63178 Aubière, France.,Centre de Recherche en Nutrition Humaine d'Auvergne, F-63000 Clermont-Ferrand, France
| | - Silvère Baron
- INSERM U 1103, Laboratoire GReD, Campus Universitaire des Cézeaux, TSA 60026, CS 60026, 63178 Aubière Cedex, France.,Université Clermont Auvergne, Université Blaise Pascal, GReD, F-63178 Aubière, France.,CNRS, UMR 6293, GReD, F-63178 Aubière, France.,Centre de Recherche en Nutrition Humaine d'Auvergne, F-63000 Clermont-Ferrand, France
| | - Kristina Schoonjans
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Françoise Caira
- INSERM U 1103, Laboratoire GReD, Campus Universitaire des Cézeaux, TSA 60026, CS 60026, 63178 Aubière Cedex, France.,Université Clermont Auvergne, Université Blaise Pascal, GReD, F-63178 Aubière, France.,CNRS, UMR 6293, GReD, F-63178 Aubière, France.,Centre de Recherche en Nutrition Humaine d'Auvergne, F-63000 Clermont-Ferrand, France
| | - David H Volle
- INSERM U 1103, Laboratoire GReD, Campus Universitaire des Cézeaux, TSA 60026, CS 60026, 63178 Aubière Cedex, France.,Université Clermont Auvergne, Université Blaise Pascal, GReD, F-63178 Aubière, France.,CNRS, UMR 6293, GReD, F-63178 Aubière, France.,Centre de Recherche en Nutrition Humaine d'Auvergne, F-63000 Clermont-Ferrand, France
| |
Collapse
|
14
|
Martinot E, Sèdes L, Baptissart M, Holota H, Rouaisnel B, Damon-Soubeyrand C, De Haze A, Saru JP, Thibault-Carpentier C, Keime C, Lobaccaro JMA, Baron S, Benoit G, Caira F, Beaudoin C, Volle DH. The Bile Acid Nuclear Receptor FXRα Is a Critical Regulator of Mouse Germ Cell Fate. Stem Cell Reports 2017; 9:315-328. [PMID: 28669602 PMCID: PMC5511114 DOI: 10.1016/j.stemcr.2017.05.036] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 05/31/2017] [Accepted: 05/31/2017] [Indexed: 12/14/2022] Open
Abstract
Spermatogenesis is the process by which spermatozoa are generated from spermatogonia. This cell population is heterogeneous, with self-renewing spermatogonial stem cells (SSCs) and progenitor spermatogonia that will continue on a path of differentiation. Only SSCs have the ability to regenerate and sustain spermatogenesis. This makes the testis a good model to investigate stem cell biology. The Farnesoid X Receptor alpha (FXRα) was recently shown to be expressed in the testis. However, its global impact on germ cell homeostasis has not yet been studied. Here, using a phenotyping approach in Fxrα−/− mice, we describe unexpected roles of FXRα on germ cell physiology independent of its effects on somatic cells. FXRα helps establish and maintain an undifferentiated germ cell pool and in turn influences male fertility. FXRα regulates the expression of several pluripotency factors. Among these, in vitro approaches show that FXRα controls the expression of the pluripotency marker Lin28 in the germ cells. FXRα regulated germ cell apoptotis independently of androgen homeostasis FXRα controls germ cell differentiation FXRα regulates the establishment and maintenance of undifferentiated germ cells In germ cells, FXRα controls the expression of pluripotency markers such as Lin28
Collapse
Affiliation(s)
- Emmanuelle Martinot
- INSERM U 1103, Université Clermont Auvergne, CNRS UMR 6293, Laboratoire GReD, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Lauriane Sèdes
- INSERM U 1103, Université Clermont Auvergne, CNRS UMR 6293, Laboratoire GReD, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Marine Baptissart
- INSERM U 1103, Université Clermont Auvergne, CNRS UMR 6293, Laboratoire GReD, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Hélène Holota
- INSERM U 1103, Université Clermont Auvergne, CNRS UMR 6293, Laboratoire GReD, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Betty Rouaisnel
- INSERM U 1103, Université Clermont Auvergne, CNRS UMR 6293, Laboratoire GReD, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Christelle Damon-Soubeyrand
- INSERM U 1103, Université Clermont Auvergne, CNRS UMR 6293, Laboratoire GReD, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France; Centre de Recherche en Nutrition Humaine d'Auvergne, 63000 Clermont-Ferrand, France
| | - Angélique De Haze
- INSERM U 1103, Université Clermont Auvergne, CNRS UMR 6293, Laboratoire GReD, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France; Centre de Recherche en Nutrition Humaine d'Auvergne, 63000 Clermont-Ferrand, France
| | - Jean-Paul Saru
- INSERM U 1103, Université Clermont Auvergne, CNRS UMR 6293, Laboratoire GReD, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France; Centre de Recherche en Nutrition Humaine d'Auvergne, 63000 Clermont-Ferrand, France
| | | | - Céline Keime
- IGBMC - CNRS UMR 7104 - Inserm U 964, 1 BP 10142, 67404 Illkirch Cedex, France
| | - Jean-Marc A Lobaccaro
- INSERM U 1103, Université Clermont Auvergne, CNRS UMR 6293, Laboratoire GReD, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France; Centre de Recherche en Nutrition Humaine d'Auvergne, 63000 Clermont-Ferrand, France
| | - Silvère Baron
- INSERM U 1103, Université Clermont Auvergne, CNRS UMR 6293, Laboratoire GReD, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France; Centre de Recherche en Nutrition Humaine d'Auvergne, 63000 Clermont-Ferrand, France
| | - Gérard Benoit
- Laboratoire de Biologie Moléculaire de la Cellule, Ecole normale supérieure de Lyon, UMR5239 CNRS/ENS Lyon/UCBL/HCL, 46, allée d'Italie, 69364 Lyon Cedex 07, France
| | - Françoise Caira
- INSERM U 1103, Université Clermont Auvergne, CNRS UMR 6293, Laboratoire GReD, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Claude Beaudoin
- INSERM U 1103, Université Clermont Auvergne, CNRS UMR 6293, Laboratoire GReD, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - David H Volle
- INSERM U 1103, Université Clermont Auvergne, CNRS UMR 6293, Laboratoire GReD, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France.
| |
Collapse
|
15
|
Kübeck R, Bonet-Ripoll C, Hoffmann C, Walker A, Müller VM, Schüppel VL, Lagkouvardos I, Scholz B, Engel KH, Daniel H, Schmitt-Kopplin P, Haller D, Clavel T, Klingenspor M. Dietary fat and gut microbiota interactions determine diet-induced obesity in mice. Mol Metab 2016; 5:1162-1174. [PMID: 27900259 PMCID: PMC5123202 DOI: 10.1016/j.molmet.2016.10.001] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 09/26/2016] [Accepted: 10/04/2016] [Indexed: 12/19/2022] Open
Abstract
Objective Gut microbiota may promote positive energy balance; however, germfree mice can be either resistant or susceptible to diet-induced obesity (DIO) depending on the type of dietary intervention. We here sought to identify the dietary constituents that determine the susceptibility to body fat accretion in germfree (GF) mice. Methods GF and specific pathogen free (SPF) male C57BL/6N mice were fed high-fat diets either based on lard or palm oil for 4 wks. Mice were metabolically characterized at the end of the feeding trial. FT-ICR-MS and UPLC-TOF-MS were used for cecal as well as hepatic metabolite profiling and cecal bile acids quantification, respectively. Hepatic gene expression was examined by qRT-PCR and cecal gut microbiota of SPF mice was analyzed by high-throughput 16S rRNA gene sequencing. Results GF mice, but not SPF mice, were completely DIO resistant when fed a cholesterol-rich lard-based high-fat diet, whereas on a cholesterol-free palm oil-based high-fat diet, DIO was independent of gut microbiota. In GF lard-fed mice, DIO resistance was conveyed by increased energy expenditure, preferential carbohydrate oxidation, and increased fecal fat and energy excretion. Cecal metabolite profiling revealed a shift in bile acid and steroid metabolites in these lean mice, with a significant rise in 17β-estradiol, which is known to stimulate energy expenditure and interfere with bile acid metabolism. Decreased cecal bile acid levels were associated with decreased hepatic expression of genes involved in bile acid synthesis. These metabolic adaptations were largely attenuated in GF mice fed the palm-oil based high-fat diet. We propose that an interaction of gut microbiota and cholesterol metabolism is essential for fat accretion in normal SPF mice fed cholesterol-rich lard as the main dietary fat source. This is supported by a positive correlation between bile acid levels and specific bacteria of the order Clostridiales (phylum Firmicutes) as a characteristic feature of normal SPF mice fed lard. Conclusions In conclusion, our study identified dietary cholesterol as a candidate ingredient affecting the crosstalk between gut microbiota and host metabolism. Cholesterol-based but not plant sterol-based high-fat diet protects germfree (GF) mice from diet-induced obesity (DIO). DIO resistant GF mice show preferential carbohydrate oxidation, higher energy expenditure and energy and fat excretion. DIO resistance in GF mice is accompanied by increased steroid hormone levels but decreased bile acid levels in the cecum. Substrate oxidation and fat excretion in DIO resistant GF mice is linked to decreased hepatic Cyp7a1 and Nr1h4 expression.
Collapse
Key Words
- ANOVA, analysis of variance
- Abcg5, ATP-binding cassette sub-family G member 5
- Abcg8, ATP-binding cassette sub-family G member 8
- Actb, beta actin
- Akr1d1, aldo-keto-reductase family member 1
- BMR, basal metabolic rate
- CA, cholic acid
- CD, control diet
- CDCA, chenodeoxycholic acid
- CIDEA, cell death inducing DFFA-like effector
- COX4, cytochrome c oxidase subunit 4
- Cyp27a1, cholesterol 27 alpha-hydroxylase
- Cyp7a1, cholesterol 7 alpha-hydroxylase
- DCA, deoxycholic acid
- DEE, daily energy expenditure
- DIO, diet-induced obesity
- Dhcr7, 7-dehydrocholesterol reductase
- Diet-induced obesity resistance
- Eef2, eukaryotic elongation factor 2
- Energy balance
- FT-ICR-MS, Fourier transform-Ion Cyclotron Resonance-Mass Spectrometry
- FT-IR, Fourier transform-infrared spectroscopy
- GF, germfree
- GUSB, beta-glucuronidase
- Germfree
- HDCA, hyodeoxycholic acid
- HP, heat production
- High-fat diet
- Hmgcr, 3-hydroxy-3-methylglutaryl Coenzyme A reductase
- Hmgcs, 3-hydroxy-3-methylglutaryl Coenzyme A synthase 1
- Hprt1, hypoxanthine guanine phosphoribosyl transferase
- Hsd11b1, hydroxysteroid (11-β) dehydrogenase 1
- Hsp90, heat shock protein 90
- LHFD, high-fat diet based on lard
- Ldlr, low density lipoprotein receptor
- MCA, muricholic acid
- Nr1h2, nuclear receptor subfamily 1, group H, member 2 (liver X receptor β)
- Nr1h3, nuclear receptor subfamily 1, group H, member 3 (liver X receptor α)
- Nr1h4, nuclear receptor subfamily 1, group H, member 4 (farnesoid X receptor α)
- PHFD, high-fat diet based on palm oil
- PRDM16, PR domain containing 16
- SPF, specific pathogen free
- Srebf1, sterol regulatory element binding transcription factor 1
- TCA, taurocholic acid
- TMCA, Tauromuricholic acid
- Tf2b, transcription factor II B
- UCP1, uncoupling protein 1
- UDCA, ursodeoxycholic acid
- UPLC-TOF-MS, ultraperformance liquid chromatography-time of flight-mass spectrometry
- qPCR, quantitative real-time polymerase chain reaction
Collapse
Affiliation(s)
- Raphaela Kübeck
- ZIEL - Institute for Food and Health, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany; Chair of Molecular Nutritional Medicine, Technical University of Munich, TUM School of Life Sciences Weihenstephan, EKFZ - Else Kröner-Fresenius-Center for Nutritional Medicine, Gregor-Mendel-Str. 2, 85354 Freising, Germany
| | - Catalina Bonet-Ripoll
- ZIEL - Institute for Food and Health, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany; Chair of Molecular Nutritional Medicine, Technical University of Munich, TUM School of Life Sciences Weihenstephan, EKFZ - Else Kröner-Fresenius-Center for Nutritional Medicine, Gregor-Mendel-Str. 2, 85354 Freising, Germany
| | - Christina Hoffmann
- ZIEL - Institute for Food and Health, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany; Chair of Molecular Nutritional Medicine, Technical University of Munich, TUM School of Life Sciences Weihenstephan, EKFZ - Else Kröner-Fresenius-Center for Nutritional Medicine, Gregor-Mendel-Str. 2, 85354 Freising, Germany
| | - Alesia Walker
- Research Unit Analytical BioGeoChemistry, Department of Environmental Sciences, Helmholtz Zentrum München, Ingolstädter Landstr.1, 85764 Neuherberg, Germany
| | - Veronika Maria Müller
- ZIEL - Institute for Food and Health, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany; Chair of Nutritional Physiology, Technical University of Munich, TUM School of Life Sciences Weihenstephan, Gregor-Mendel-Str. 2, 85354 Freising, Germany
| | - Valentina Luise Schüppel
- ZIEL - Institute for Food and Health, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany; Chair of Nutrition and Immunology, Technical University of Munich, TUM School of Life Sciences Weihenstephan, Maximus-von-Imhof-Forum 2, 85354 Freising, Germany
| | - Ilias Lagkouvardos
- ZIEL - Institute for Food and Health, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany
| | - Birgit Scholz
- Chair of General Food Technology, Technische Universität München, Alte Akademie 10, 85354 Freising, Germany
| | - Karl-Heinz Engel
- Chair of General Food Technology, Technische Universität München, Alte Akademie 10, 85354 Freising, Germany
| | - Hannelore Daniel
- Chair of Nutritional Physiology, Technical University of Munich, TUM School of Life Sciences Weihenstephan, Gregor-Mendel-Str. 2, 85354 Freising, Germany
| | - Philippe Schmitt-Kopplin
- ZIEL - Institute for Food and Health, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany; Research Unit Analytical BioGeoChemistry, Department of Environmental Sciences, Helmholtz Zentrum München, Ingolstädter Landstr.1, 85764 Neuherberg, Germany; Chair of Analytical Food Chemistry, Technische Universität München, Alte Akademie 10, 85354 Freising, Germany
| | - Dirk Haller
- ZIEL - Institute for Food and Health, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany; Chair of Nutrition and Immunology, Technical University of Munich, TUM School of Life Sciences Weihenstephan, Maximus-von-Imhof-Forum 2, 85354 Freising, Germany
| | - Thomas Clavel
- ZIEL - Institute for Food and Health, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany
| | - Martin Klingenspor
- ZIEL - Institute for Food and Health, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany; Chair of Molecular Nutritional Medicine, Technical University of Munich, TUM School of Life Sciences Weihenstephan, EKFZ - Else Kröner-Fresenius-Center for Nutritional Medicine, Gregor-Mendel-Str. 2, 85354 Freising, Germany.
| |
Collapse
|
16
|
Qin X, Liu M, Wang X. New insights into the androgen biotransformation in prostate cancer: A regulatory network among androgen, androgen receptors and UGTs. Pharmacol Res 2016; 106:114-122. [PMID: 26926093 DOI: 10.1016/j.phrs.2016.02.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 02/22/2016] [Accepted: 02/23/2016] [Indexed: 01/15/2023]
Abstract
Androgen, as one kind of steroid hormones, is pivotal in the hormone-sensitive cancer, such as prostate cancer (PCa). The synthesis, elimination, and bioavailability of androgen in prostate cells have been proved to be a main cause of the carcinogenesis, maintenance and deterioration of PCa. This review illustrates the outlines of androgen biotransformation, and further discusses the different enzymes, especially UDP-glucuronyltransferases (UGTs) embedded in both benign and malignant prostate cells, which catalyze the reactions. Although many inhibitors of the enzymes responsible for the synthesis of androgens have been developed into drugs to fight against PCa, the elimination procedures metabolized by the UGTs are less emphasized. Thus the regulatory network among androgen, androgen receptors (AR) and UGTs is carefully reviewed in this article, indicating the determinant effects of UGTs on prostatic androgens and the regulation of AR. Finally, the hypothesis is also put forward that the regulators of UGTs may be developed to accelerate the androgen elimination and benefit PCa therapy.
Collapse
Affiliation(s)
- Xuan Qin
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China; Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Houston, TX, USA
| | - Xin Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China.
| |
Collapse
|
17
|
Zhang HM, Wang X, Wu ZH, Liu HL, Chen W, Zhang ZZ, Chen D, Zeng TS. Beneficial effect of farnesoid X receptor activation on metabolism in a diabetic rat model. Mol Med Rep 2016; 13:2135-42. [PMID: 26782298 DOI: 10.3892/mmr.2016.4761] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 11/17/2015] [Indexed: 11/06/2022] Open
Abstract
Farnesoid X receptor (FXR) is an important regulator of glucose and lipid homeostasis. However, the exact role of FXR in diabetes remains to be fully elucidated. The present study examined the effects of chenodeoxycholic acid (CDCA), an agonist of FXR, on metabolism profile in a rat model of type 2 diabetes mellitus (T2DM). Male Wistar rats (8‑week‑old; n=40) were randomized into the following four groups (n=10): Untreated control, CDCA‑treated, T2DM, and CDCA‑treated T2DM. To establish the T2DM model, the rats were fed a high‑fat diet (HFD) for 4 weeks and received a single low‑dose intraperitoneal injection of streptozotocin (30 mg/kg), followed by an additional 4 weeks of HFD feeding. CDCA was administrated (10 mg/kg/d) intraperitoneally for 10 days. Reverse transcription‑quantitative polymerase chain reaction and western blotting assays were performed to determine the RNA and protein expression of FXR, phosphoenolpyruvate carboxykinase, G6Pase, proliferator‑activated receptor‑γ coactivator‑1 and short heterodimer partner in rat liver tissue. The results revealed that FXR activation by CDCA did not reduce body weight, but it lowered the plasma levels of fasting glucose, insulin and triglycerides in the T2DM rats. CDCA administration reversed the downregulation of the mRNA and protein expression of FXR in the T2DM rat liver tissue samples. Furthermore, treatment with CDCA reduced the mRNA and protein expression levels of phosphoenolpyruvate carboxykinase, glucose 6‑phosphatase and peroxisome proliferator‑activated receptor‑γ coactivator‑1 in the liver tissue samples of the T2DM rats. By contrast, CDCA treatment increased the mRNA and protein expression levels of short heterodimer partner in the liver tissue samples of the T2DM rats. In conclusion, FXR agonist treatment induces beneficial effects on metabolism in the rat T2DM model. In conclusion, the present study indicated that the FXR agonist may be useful for the treatment of T2DM and hypertriglyceridemia.
Collapse
Affiliation(s)
- Hong-Mei Zhang
- Department of Endocrinology, Hubei Integrated Traditional Chinese and Western Medicine Hospital, Wuhan, Hubei 430015, P.R. China
| | - Xuan Wang
- Department of Clinical Teaching and Research, College of Health Science and Nursing, Wuhan Polytechnic University, Wuhan, Hubei 430015, P.R. China
| | - Zhao-Hong Wu
- Department of Endocrinology, Hubei Integrated Traditional Chinese and Western Medicine Hospital, Wuhan, Hubei 430015, P.R. China
| | - Hui-Ling Liu
- Department of Endocrinology, Hubei Integrated Traditional Chinese and Western Medicine Hospital, Wuhan, Hubei 430015, P.R. China
| | - Wei Chen
- Department of Endocrinology, Hubei Integrated Traditional Chinese and Western Medicine Hospital, Wuhan, Hubei 430015, P.R. China
| | - Zhong-Zhi Zhang
- Department of Endocrinology, Hubei Integrated Traditional Chinese and Western Medicine Hospital, Wuhan, Hubei 430015, P.R. China
| | - Dan Chen
- Department of Endocrinology, Hubei Integrated Traditional Chinese and Western Medicine Hospital, Wuhan, Hubei 430015, P.R. China
| | - Tian-Shu Zeng
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
18
|
Evaluation and identification of dioxin exposure biomarkers in human urine by high-resolution metabolomics, multivariate analysis and in vitro synthesis. Toxicol Lett 2016; 240:22-31. [DOI: 10.1016/j.toxlet.2015.10.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 10/06/2015] [Accepted: 10/08/2015] [Indexed: 12/13/2022]
|
19
|
Bile Acid Alters Male Mouse Fertility in Metabolic Syndrome Context. PLoS One 2015; 10:e0139946. [PMID: 26439743 PMCID: PMC4595338 DOI: 10.1371/journal.pone.0139946] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 09/18/2015] [Indexed: 12/25/2022] Open
Abstract
Bile acids have recently been demonstrated as molecules with endocrine activities controlling several physiological functions such as immunity and glucose homeostases. They act mainly through two receptors, the nuclear receptor Farnesol-X-Receptor alpha (FXRα) and the G-protein coupled receptor (TGR5). These recent studies have led to the idea that molecules derived from bile acids (BAs) and targeting their receptors must be good targets for treatment of metabolic diseases such as obesity or diabetes. Thus it might be important to decipher the potential long term impact of such treatment on different physiological functions. Indeed, BAs have recently been demonstrated to alter male fertility. Here we demonstrate that in mice with overweight induced by high fat diet, BA exposure leads to increased rate of male infertility. This is associated with the altered germ cell proliferation, default of testicular endocrine function and abnormalities in cell-cell interaction within the seminiferous epithelium. Even if the identification of the exact molecular mechanisms will need more studies, the present results suggest that both FXRα and TGR5 might be involved. We believed that this work is of particular interest regarding the potential consequences on future approaches for the treatment of metabolic diseases.
Collapse
|
20
|
Bile acid signaling through farnesoid X and TGR5 receptors in hepatobiliary and intestinal diseases. Hepatobiliary Pancreat Dis Int 2015; 14:18-33. [PMID: 25655287 DOI: 10.1016/s1499-3872(14)60307-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND The well-known functions of bile acids (BAs) are the emulsification and absorption of lipophilic xenobiotics. However, the emerging evidences in the past decade showed that BAs act as signaling molecules that not only autoregulate their own metabolism and enterohepatic recirculation, but also as important regulators of integrative metabolism by activating nuclear and membrane-bound G protein-coupled receptors. The present review was to get insight into the role of maintenance of BA homeostasis and BA signaling pathways in development and management of hepatobiliary and intestinal diseases. DATA SOURCES Detailed and comprehensive search of PubMed and Scopus databases was carried out for original and review articles. RESULTS Disturbances in BA homeostasis contribute to the development of several hepatobiliary and intestinal disorders, such as non-alcoholic fatty liver disease, liver cirrhosis, cholesterol gallstone disease, intestinal diseases and both hepatocellular and colorectal carcinoma. CONCLUSION Further efforts made in order to advance the understanding of sophisticated BA signaling network may be promising in developing novel therapeutic strategies related not only to hepatobiliary and gastrointestinal but also systemic diseases.
Collapse
|
21
|
Anaya-Hernández A, Méndez-Tepepa M, Laura G. HA, Pacheco P, Martínez-Gómez M, Castelán F, Cuevas E. Farnesoid X receptor immunolocalization in reproductive tissues of adult female rabbits. Acta Histochem 2014; 116:1068-74. [PMID: 24975630 DOI: 10.1016/j.acthis.2014.05.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 05/02/2014] [Accepted: 05/04/2014] [Indexed: 12/21/2022]
Abstract
Farnesoid X receptor (FXR) has been involved in lipid metabolism, cell proliferation, apoptosis, and aromatase expression, as well as in the steroid synthesis and signaling. Considering that these events occur in reproductive tissues in females, the aim of the present study was to determine the immunolocalization of FXR in the ovary, oviduct, uterus, and vagina of rabbits. Rabbits were sacrificed and their reproductive tissues were excised and histologically processed. Immunohistochemistry for FXR was done and reproductive tissues were photographed. FXR immunoreactivity was found in all types of ovarian follicles, ovarian stroma, and corpus luteum of virgin and pregnant rabbits. Also, oviductal and vaginal epithelium of virgins, as well as the oviductal smooth muscle, showed anti-FXR immunoreactivity. The uterine epithelium and musculature of virgins had scarce anti-FXR immunoreactivity. Although the role of FXR in female reproductive tissues is still not known, it is possible to consider various functions related to the reproductive tissue.
Collapse
|
22
|
Sanchez R, Schuermann Y, Gagnon-Duval L, Baldassarre H, Murphy BD, Gevry N, Agellon LB, Bordignon V, Duggavathi R. Differential abundance of IGF1, bile acids, and the genes involved in their signaling in the dominant follicle microenvironment of lactating cows and nulliparous heifers. Theriogenology 2014; 81:771-9. [PMID: 24503106 DOI: 10.1016/j.theriogenology.2014.01.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 12/29/2013] [Accepted: 01/01/2014] [Indexed: 12/29/2022]
Abstract
It is well documented that incidence of fertility problems is high in lactating cows but not in heifers of the same genetic merit. Understanding the metabolic and molecular differences between fertile heifers and relatively infertile lactating cows will help us understand the pathogenesis of infertility in dairy cows. Follicular waves in lactating cows (30-50 days in milk; n = 12) and heifers (n = 10) were synchronized by ultrasound-guided follicle ablation. Follicular fluid and granulosa cells of the dominant follicle were collected by ultrasound-guided aspiration along with blood sampling on Day 6 after synchronization. Dominant and subordinate follicles were larger in lactating cows than in heifers. Metabolic stress in lactating cows was evidenced by lower glucose and higher ß-hydroxy butyric acid compared with heifers. Insulin-like growth factor 1 signaling was reduced in the dominant follicle in lactating cows through reduced insulin-like growth factor 1 concentrations in plasma and follicular fluid of the dominant follicle, and reduced expression of pregnancy-associated plasma protein A (PAPPA) in their granulosa cells. We also found increased levels of total bile acids in the follicular fluid of the dominant follicle of lactating cows compared with heifers. Granulosa cells of the dominant follicle had higher expression of SLC10A2 and GPBAR1 (bile acid transporter and receptor, respectively) in lactating cows. These novel data are indicative of increased bile acid signaling within the dominant follicles of lactating cows compared with heifers. Overall, we demonstrate in the present study the metabolic, endocrine, and molecular differences within the microenvironment of the dominant follicles in lactating cows and heifers. These differences in follicular microenvironment may contribute toward abnormal ovarian function in lactating dairy cows.
Collapse
Affiliation(s)
- Ricardo Sanchez
- Department of Animal Science, McGill University, Ste-Anne-de-Bellevue, Quebec, Canada
| | - Yasmin Schuermann
- Department of Animal Science, McGill University, Ste-Anne-de-Bellevue, Quebec, Canada
| | - Laurianne Gagnon-Duval
- Centre de recherche en reproduction animale, Université de Montréal, St-Hyacinthe, Quebec, Canada
| | - Hernan Baldassarre
- Department of Animal Science, McGill University, Ste-Anne-de-Bellevue, Quebec, Canada
| | - Bruce D Murphy
- Centre de recherche en reproduction animale, Université de Montréal, St-Hyacinthe, Quebec, Canada
| | - Nicolas Gevry
- Département de Biologie, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Luis B Agellon
- School of Dietetics and Human Nutrition, McGill University, Ste-Anne-de-Bellevue, Quebec, Canada
| | - Vilceu Bordignon
- Department of Animal Science, McGill University, Ste-Anne-de-Bellevue, Quebec, Canada
| | - Raj Duggavathi
- Department of Animal Science, McGill University, Ste-Anne-de-Bellevue, Quebec, Canada.
| |
Collapse
|
23
|
Long MD, Thorne JL, Russell J, Battaglia S, Singh PK, Sucheston-Campbell LE, Campbell MJ. Cooperative behavior of the nuclear receptor superfamily and its deregulation in prostate cancer. Carcinogenesis 2013; 35:262-71. [PMID: 24104552 DOI: 10.1093/carcin/bgt334] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The current study aimed to assess the topology of the nuclear receptor (NR) superfamily in normal prostate epithelial cells and its distortion in prostate cancer. Both in vitro and in silico approaches were utilized to profile NRs expressed in non-malignant RWPE-1 cells, which were subsequently investigated by treating cells with 132 binary NR ligand combinations. Nine significant cooperative interactions emerged including both superadditive [22(R)-hydroxycholesterol and eicosatetraenoic acid] and subadditive [1α,25(OH)2D3 and chenodeoxycholic acid] cellular responses, which could be explained in part by cooperative control of cell-cycle progression and candidate gene expression. In addition, publicly available data were employed to assess NR expression in human prostate tissue. Common and significant loss of NR superfamily expression was established in publicly available data from prostate tumors, in part predicting parallel distortion of targeting microRNA. These findings suggest that the NR superfamily in the prostate cooperatively integrates signals from dietary, hormonal and metabolic cues, and is significantly distorted in prostate cancer.
Collapse
Affiliation(s)
- Mark D Long
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | | | | | | | | | | | | |
Collapse
|