1
|
Høgh S, Borgsted C, Hegaard HK, Renault KM, Ekelund K, Bruzzone SE, Clemmensen C, Klein AB, Frokjaer VG. Growth Differentiation Factor 15 during pregnancy and postpartum as captured in blood, cerebrospinal fluid and placenta: A cohort study on associations with maternal mental health. Psychoneuroendocrinology 2025; 171:107212. [PMID: 39418693 DOI: 10.1016/j.psyneuen.2024.107212] [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: 06/21/2024] [Revised: 09/20/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024]
Abstract
INTRODUCTION Growth Differentiation Factor 15 (GDF15) increases substantially during pregnancy and is primarily produced by the placenta. Elevated levels of GDF15 have been associated with mental health problems in non-perinatal populations, higher corticosterone levels, and decreased estrogen receptor activity. However, the role of GDF15 in mental health during the perinatal transition remains unknown. This longitudinal study is the first to evaluate pregnancy levels of GDF15 in cerebrospinal fluid (cGDF15), plasma (pGDF15) and placenta GDF15 mRNA, along with mapping plasma GDF15 (pGDF15) level changes from late pregnancy to early postpartum. Moreover, we aimed to evaluate the association between pregnancy cGDF15 levels and cortisol early postpartum, evaluate the association between pregnancy cGDF15 levels and mental health in pregnancy and postpartum, and evaluate the association between pGDF15 and estrogens and high-sensitivity C-reactive protein (CRP). METHODS We included data from 95 women scheduled for a planned cesarean section and obtained cerebrospinal fluid (CSF) and plasma levels of GDF15. We quantified GDF15 mRNA levels in placenta biopsies. Estrogens, high-sensitivity CRP, and mental health measures were further collected on the day or one day before the cesarean section. At five weeks postpartum, mental health measures and saliva samples for cortisol analyses were collected. Correlation analyses for GDF15 in CSF, plasma, and placenta mRNA were performed, along with association analyses for pregnancy cGDF15, Cortisol Awakening Response, and mental health outcomes. RESULTS We demonstrated a strong correlation between cGDF15 and pGDF15 (r=0.52; p<0.001) and found that both cGDF15 and pGDF15 correlated with placenta GDF15 mRNA*placental weight (r=0.62, p<0.001 and r=0.44, p=0.008, respectively). During late pregnancy, both estradiol (E2) and estriol (E3) were significantly associated with pGDF15 levels (E2: p=0.002; E3: p(corrected)<0.001). Finally, we found that cGDF15 levels were not associated with self-reported mental well-being or the Cortisol Awakening Response or absolute cortisol at awakening postpartum. CONCLUSION This novel study points to the unique hormonal landscape during the perinatal transition and the specific role of GDF15 in pregnancy, which appears uncoupled with perinatal mental health and cortisol outcomes. Our data also strongly imply that the overall amount of circulating GDF15 in late pregnancy is closely related to placenta size.
Collapse
Affiliation(s)
- Stinne Høgh
- Neurobiology Research Unit, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark; Department of Obstetrics, Copenhagen University Hospital - Rigshospitalet, Denmark
| | - Camilla Borgsted
- Neurobiology Research Unit, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; Mental Health Center Copenhagen, Copenhagen University Hospital, Denmark
| | - Hanne K Hegaard
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark; Department of Obstetrics, Copenhagen University Hospital - Rigshospitalet, Denmark
| | - Kristina M Renault
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark; Department of Obstetrics, Copenhagen University Hospital - Rigshospitalet, Denmark
| | - Kim Ekelund
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark; Department of Anaesthesiology, Copenhagen University Hospital - Rigshospitalet, Denmark
| | - Silvia Ep Bruzzone
- Neurobiology Research Unit, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Christoffer Clemmensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anders B Klein
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Vibe G Frokjaer
- Neurobiology Research Unit, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark; Mental Health Center Copenhagen, Copenhagen University Hospital, Denmark.
| |
Collapse
|
2
|
Burra P, Zanetto A, Schnabl B, Reiberger T, Montano-Loza AJ, Asselta R, Karlsen TH, Tacke F. Hepatic immune regulation and sex disparities. Nat Rev Gastroenterol Hepatol 2024; 21:869-884. [PMID: 39237606 DOI: 10.1038/s41575-024-00974-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/25/2024] [Indexed: 09/07/2024]
Abstract
Chronic liver disease is a major cause of morbidity and mortality worldwide. Epidemiology, clinical phenotype and response to therapies for gastrointestinal and liver diseases are commonly different between women and men due to sex-specific hormonal, genetic and immune-related factors. The hepatic immune system has unique regulatory functions that promote the induction of intrahepatic tolerance, which is key for maintaining liver health and homeostasis. In liver diseases, hepatic immune alterations are increasingly recognized as a main cofactor responsible for the development and progression of chronic liver injury and fibrosis. In this Review, we discuss the basic mechanisms of sex disparity in hepatic immune regulation and how these mechanisms influence and modify the development of autoimmune liver diseases, genetic liver diseases, portal hypertension and inflammation in chronic liver disease. Alterations in gut microbiota and their crosstalk with the hepatic immune system might affect the progression of liver disease in a sex-specific manner, creating potential opportunities for novel diagnostic and therapeutic approaches to be evaluated in clinical trials. Finally, we identify and propose areas for future basic, translational and clinical research that will advance our understanding of sex disparities in hepatic immunity and liver disease.
Collapse
Affiliation(s)
- Patrizia Burra
- Gastroenterology and Multivisceral Transplant Unit, Department of Surgery, Oncology, and Gastroenterology, Padua University Hospital, Padua, Italy.
| | - Alberto Zanetto
- Gastroenterology and Multivisceral Transplant Unit, Department of Surgery, Oncology, and Gastroenterology, Padua University Hospital, Padua, Italy
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Medicine, VA San Diego Healthcare System, San Diego, CA, USA
| | - Thomas Reiberger
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Aldo J Montano-Loza
- Division of Gastroenterology and Liver Unit, Department of Medicine, University of Alberta Hospital, Edmonton, Alberta, Canada
| | - Rosanna Asselta
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Tom Hemming Karlsen
- Department of Transplantation Medicine, Clinic of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital and University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Clinic of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| |
Collapse
|
3
|
Cherubini A, Della Torre S, Pelusi S, Valenti L. Sexual dimorphism of metabolic dysfunction-associated steatotic liver disease. Trends Mol Med 2024; 30:1126-1136. [PMID: 38890029 DOI: 10.1016/j.molmed.2024.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 06/20/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common chronic liver condition. MASLD is a sexually dimorphic condition, with its development and progression influenced by sex chromosomes and hormones. Estrogens typically protect against, whereas androgens promote, MASLD. Therapeutic approaches for a sex-specific personalized medicine include estrogen replacement, androgen blockers, and novel drugs targeting hormonal pathways. However, the interactions between hormonal factors and inherited genetic variation impacts MASLD risk, necessitating more tailored therapies. Understanding sex disparities and the role of estrogens could improve MASLD interventions and management, whereas clinical trials addressing sex differences are crucial for advancing personalized treatment. This review explores the underappreciated impact of sexual dimorphism in MASLD and discusses the potential therapeutic application of sex-related hormones.
Collapse
Affiliation(s)
- Alessandro Cherubini
- Department of Transfusion Medicine, Precision Medicine Lab, Biological Resource Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Sara Della Torre
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| | - Serena Pelusi
- Department of Transfusion Medicine, Precision Medicine Lab, Biological Resource Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Luca Valenti
- Department of Transfusion Medicine, Precision Medicine Lab, Biological Resource Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
4
|
Labour A, Lac M, Frassin L, Lair B, Murphy E, Maslo C, Monbrun L, Calmy ML, Marquès M, Viguerie N, Tavernier G, Gourdy P, O'Gorman D, Montastier E, Laurens C, Montagner A, Moro C. GDF15 is dispensable for the insulin-sensitizing effects of chronic exercise. Cell Rep 2024; 43:114577. [PMID: 39096490 DOI: 10.1016/j.celrep.2024.114577] [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: 01/04/2024] [Revised: 06/28/2024] [Accepted: 07/18/2024] [Indexed: 08/05/2024] Open
Abstract
Growth and differentiation factor 15 (GDF15) has recently emerged as a weight loss and insulin-sensitizing factor. Growing evidence also supports a role for GDF15 as a physiological, exercise-induced stress signal. Here, we tested whether GDF15 is required for the insulin-sensitizing effects of exercise in mice and humans. At baseline, both under a standard nutritional state and high-fat feeding, GDF15 knockout (KO) mice display normal glucose tolerance, systemic insulin sensitivity, maximal speed, and endurance running capacity when compared to wild-type littermates independent of sex. When submitted to a 4-week exercise training program, both lean and obese wild-type and GDF15 KO mice similarly improve their endurance running capacity, glucose tolerance, systemic insulin sensitivity, and peripheral glucose uptake. Insulin-sensitizing effects of exercise training were also unrelated to changes in plasma GDF15 in humans. In summary, we here show that GDF15 is dispensable for the insulin-sensitizing effects of chronic exercise.
Collapse
Affiliation(s)
- Axel Labour
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Toulouse, France
| | - Marlène Lac
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Toulouse, France
| | - Lucas Frassin
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Toulouse, France
| | - Benjamin Lair
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Toulouse, France
| | - Enda Murphy
- School of Health and Human Performance, Dublin City University, Dublin, Ireland
| | - Claire Maslo
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Toulouse, France
| | - Laurent Monbrun
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Toulouse, France
| | - Marie-Lou Calmy
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Toulouse, France
| | - Marie Marquès
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Toulouse, France
| | - Nathalie Viguerie
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Toulouse, France
| | - Geneviève Tavernier
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Toulouse, France
| | - Pierre Gourdy
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Toulouse, France; Department of Diabetology, Toulouse University Hospital, Toulouse, France
| | - Donal O'Gorman
- School of Health and Human Performance, Dublin City University, Dublin, Ireland
| | - Emilie Montastier
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Toulouse, France; Department of Endocrinology, Metabolic Diseases and Nutrition, Toulouse University Hospital, Toulouse, France
| | - Claire Laurens
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Toulouse, France
| | - Alexandra Montagner
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Toulouse, France
| | - Cedric Moro
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Toulouse, France.
| |
Collapse
|
5
|
Booijink R, Ramachandran P, Bansal R. Implications of innate immune sexual dimorphism for MASLD pathogenesis and treatment. Trends Pharmacol Sci 2024; 45:614-627. [PMID: 38853100 DOI: 10.1016/j.tips.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 04/18/2024] [Accepted: 05/09/2024] [Indexed: 06/11/2024]
Abstract
Growing evidence suggests that metabolic dysfunction-associated steatotic liver disease (MASLD) is significantly higher in men versus women. Increased prevalence is observed in postmenopausal women, suggesting that age and sex (hormones) influence MASLD development and progression. Molecular data further reveal that sex regulates the innate immune responses with an essential role in MASLD progression. To date, there has been limited focus on the role of innate immune sexual dimorphism in MASLD, and differences between men and women are not considered in the current drug discovery landscape. In this review, we summarize the sex disparities and innate immune sexual dimorphism in MASLD pathogenesis. We further highlight the importance of harnessing sexual dimorphism in identifying therapeutic targets, developing pharmacological therapies, and designing (pre-) clinical studies for the personalized treatment for MASLD.
Collapse
Affiliation(s)
- Richell Booijink
- Personalized Diagnostics and Therapeutics, Department of Bioengineering Technologies, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Prakash Ramachandran
- University of Edinburgh Centre for Inflammation Research, Institute for Regeneration and Repair, Edinburgh BioQuarter, Edinburgh, UK
| | - Ruchi Bansal
- Personalized Diagnostics and Therapeutics, Department of Bioengineering Technologies, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands.
| |
Collapse
|
6
|
Zhang Z, Yang Q, Jin M, Wang J, Chai Y, Zhang L, Jiang Z, Yu Q. Tamoxifen upregulates the peroxisomal β-oxidation enzyme Enoyl CoA hydratase and 3-hydroxyacyl CoA hydratase ameliorating hepatic lipid accumulation in mice. Int J Biochem Cell Biol 2024; 172:106585. [PMID: 38734232 DOI: 10.1016/j.biocel.2024.106585] [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: 10/07/2023] [Revised: 04/30/2024] [Accepted: 05/03/2024] [Indexed: 05/13/2024]
Abstract
Tamoxifen is an estrogen receptor modulator that has been reported to alleviate hepatic lipid accumulation in mice, but the mechanism is still unclear. Peroxisome fatty acid β-oxidation is the main metabolic pathway for the overload of long-chain fatty acids. As long-chain fatty acids are a cause of hepatic lipid accumulation, the activation of peroxisome fatty acid β-oxidation might be a novel therapeutic strategy for metabolic associated fatty liver disease. In this study, we investigated the mechanism of tamoxifen against hepatic lipid accumulation based on the activation of peroxisome fatty acid β-oxidation. Tamoxifen reduced liver long-chain fatty acids and relieved hepatic lipid accumulation in high fat diet mice without sex difference. In vitro, tamoxifen protected primary hepatocytes against palmitic acid-induced lipotoxicity. Mechanistically, the RNA-sequence of hepatocytes isolated from the liver revealed that peroxisome fatty acid β-oxidation was activated by tamoxifen. Protein and mRNA expression of enoyl CoA hydratase and 3-hydroxyacyl CoA hydratase were significantly increased in vivo and in vitro. Small interfering RNA enoyl CoA hydratase and 3-hydroxyacyl CoA hydratase in primary hepatocytes abolished the therapeutic effects of tamoxifen in lipid accumulation. In conclusion, our results indicated that tamoxifen could relieve hepatic lipid accumulation in high fat diet mice based on the activation of enoyl CoA hydratase and 3-hydroxyacyl CoA hydratase-mediated peroxisome fatty acids β-oxidation.
Collapse
Affiliation(s)
- Ziling Zhang
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Qinqin Yang
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Ming Jin
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Jie Wang
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Yuanyuan Chai
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Luyong Zhang
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China.
| | - Zhenzhou Jiang
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China.
| | - Qinwei Yu
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
7
|
Kumari R, Ponte ME, Franczak E, Prom JC, O'Neil MF, Sardiu ME, Lutkewitte AJ, Christenson LK, Shankar K, Morris EM, Thyfault JP. VCD-induced menopause mouse model reveals reprogramming of hepatic metabolism. Mol Metab 2024; 82:101908. [PMID: 38432400 PMCID: PMC10944007 DOI: 10.1016/j.molmet.2024.101908] [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: 12/15/2023] [Revised: 02/23/2024] [Accepted: 02/26/2024] [Indexed: 03/05/2024] Open
Abstract
OBJECTIVE Menopause adversely impacts systemic energy metabolism and increases the risk of metabolic disease(s) including hepatic steatosis, but the mechanisms are largely unknown. Dosing female mice with vinyl cyclohexene dioxide (VCD) selectively causes follicular atresia in ovaries, leading to a murine menopause-like phenotype. METHODS In this study, we treated female C57BL6/J mice with VCD (160 mg/kg i.p. for 20 consecutive days followed by verification of the lack of estrous cycling) to investigate changes in body composition, energy expenditure (EE), hepatic mitochondrial function, and hepatic steatosis across different dietary conditions. RESULTS VCD treatment induced ovarian follicular loss and increased follicle-stimulating hormone (FSH) levels in female mice, mimicking a menopause-like phenotype. VCD treatment did not affect body composition, or EE in mice on a low-fat diet (LFD) or in response to a short-term (1-week) high-fat, high sucrose diet (HFHS). However, the transition to a HFHS lowered cage activity in VCD mice. A chronic HFHS diet (16 weeks) significantly increased weight gain, fat mass, and hepatic steatosis in VCD-treated mice compared to HFHS-fed controls. In the liver, VCD mice showed suppressed hepatic mitochondrial respiration on LFD, while chronic HFHS resulted in compensatory increases in hepatic mitochondrial respiration. Also, liver RNA sequencing revealed that VCD promoted global upregulation of hepatic lipid/cholesterol synthesis pathways. CONCLUSION Our findings suggest that the VCD-induced menopause model compromises hepatic mitochondrial function and lipid/cholesterol homeostasis that sets the stage for HFHS diet-induced steatosis while also increasing susceptibility to obesity.
Collapse
Affiliation(s)
- Roshan Kumari
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA; Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, MO, USA; KU Diabetes Institute and Kansas Center for Metabolism and Obesity, University of Kansas Medical Center, Kansas City, KS, USA; Center for Children's Healthy Lifestyles and Nutrition, Kansas City, MO, USA
| | - Michael E Ponte
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA; KU Diabetes Institute and Kansas Center for Metabolism and Obesity, University of Kansas Medical Center, Kansas City, KS, USA
| | - Edziu Franczak
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA; Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, MO, USA; KU Diabetes Institute and Kansas Center for Metabolism and Obesity, University of Kansas Medical Center, Kansas City, KS, USA
| | - John C Prom
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Maura F O'Neil
- Department of Pathology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Mihaela E Sardiu
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, KS, USA; KU Diabetes Institute and Kansas Center for Metabolism and Obesity, University of Kansas Medical Center, Kansas City, KS, USA
| | - Andrew J Lutkewitte
- KU Diabetes Institute and Kansas Center for Metabolism and Obesity, University of Kansas Medical Center, Kansas City, KS, USA; Department of Internal Medicine, Division of Endocrinology, Diabetes, and Clinical Pharmacology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Lane K Christenson
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Kartik Shankar
- Department of Pediatrics, Anschutz Medical Campus, University of Colorado, Aurora, CO, USA
| | - E Matthew Morris
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA; KU Diabetes Institute and Kansas Center for Metabolism and Obesity, University of Kansas Medical Center, Kansas City, KS, USA; Center for Children's Healthy Lifestyles and Nutrition, Kansas City, MO, USA.
| | - John P Thyfault
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA; Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, MO, USA; KU Diabetes Institute and Kansas Center for Metabolism and Obesity, University of Kansas Medical Center, Kansas City, KS, USA; Department of Internal Medicine, Division of Endocrinology, Diabetes, and Clinical Pharmacology, University of Kansas Medical Center, Kansas City, KS, USA; Center for Children's Healthy Lifestyles and Nutrition, Kansas City, MO, USA.
| |
Collapse
|
8
|
Sinha RA. Targeting nuclear receptors for NASH/MASH: From bench to bedside. LIVER RESEARCH 2024; 8:34-45. [PMID: 38544909 PMCID: PMC7615772 DOI: 10.1016/j.livres.2024.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
The onset of metabolic dysfunction-associated steatohepatitis (MASH) or non-alcoholic steatohepatitis (NASH) represents a tipping point leading to liver injury and subsequent hepatic complications in the natural progression of what is now termed metabolic dysfunction-associated steatotic liver diseases (MASLD), formerly known as non-alcoholic fatty liver disease (NAFLD). With no pharmacological treatment currently available for MASH/NASH, the race is on to develop drugs targeting multiple facets of hepatic metabolism, inflammation, and pro-fibrotic events, which are major drivers of MASH. Nuclear receptors (NRs) regulate genomic transcription upon binding to lipophilic ligands and govern multiple aspects of liver metabolism and inflammation. Ligands of NRs may include hormones, lipids, bile acids, and synthetic ligands, which upon binding to NRs regulate the transcriptional activities of target genes. NR ligands are presently the most promising drug candidates expected to receive approval from the United States Food and Drug Administration as a pharmacological treatment for MASH. This review aims to cover the current understanding of NRs, including nuclear hormone receptors, non-steroid hormone receptors, circadian NRs, and orphan NRs, which are currently undergoing clinical trials for MASH treatment, along with NRs that have shown promising results in preclinical studies.
Collapse
Affiliation(s)
- Rohit A Sinha
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| |
Collapse
|
9
|
Kumari R, Ponte ME, Franczak E, Prom JC, O'Neil MF, Sardiu ME, Lutkewitte AJ, Shankar K, Morris EM, Thyfault JP. VCD-induced menopause mouse model reveals reprogramming of hepatic metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.14.571644. [PMID: 38168213 PMCID: PMC10760158 DOI: 10.1101/2023.12.14.571644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Menopause adversely impacts systemic energy metabolism and increases the risk of metabolic disease(s) including hepatic steatosis, but the mechanisms are largely unknown. Dosing female mice with vinyl cyclohexene dioxide (VCD) selectively causes follicular atresia in ovaries, leading to a murine menopause-like phenotype. In this study, we treated female C57BL6/J mice with VCD (160mg/kg i.p. for 20 consecutive days followed by verification of the lack of estrous cycling) to investigate changes in body composition, energy expenditure (EE), hepatic mitochondrial function, and hepatic steatosis across different dietary conditions. VCD treatment induced ovarian follicular loss and increased follicle-stimulating hormone (FSH) levels in female mice, mimicking a menopause-like phenotype. VCD treatment did not affect body composition, or EE in mice on a low-fat diet or in response to a short-term (1-week) high-fat, high sucrose diet (HFHS). However, the transition to a HFHS lowered cage activity in VCD mice. A chronic HFHS diet (16 weeks) significantly increased weight gain, fat mass, and hepatic steatosis in VCD-treated mice compared to HFHS-fed controls. In the liver, VCD mice showed suppressed hepatic mitochondrial respiration on LFD, while chronic HFHS diet resulted in compensatory increases in hepatic mitochondrial respiration. Also, liver RNA sequencing revealed that VCD promoted global upregulation of hepatic lipid/cholesterol synthesis pathways. Our findings suggest that the VCD- induced menopause model compromises hepatic mitochondrial function and lipid/cholesterol homeostasis that sets the stage for HFHS diet-induced steatosis while also increasing susceptibility to obesity.
Collapse
|
10
|
Dong XC, Xu DY. Research Progress on the Role and Mechanism of GDF15 in Body Weight Regulation. Obes Facts 2023; 17:1-11. [PMID: 37989122 PMCID: PMC10836939 DOI: 10.1159/000535089] [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: 04/12/2023] [Accepted: 11/06/2023] [Indexed: 11/23/2023] Open
Abstract
BACKGROUND Growth differentiation factor-15 (GDF15) is a member of the growth differentiation factor subfamily in the transforming growth factor beta superfamily. GDF15 has multiple functions and can regulate biological processes. High levels of GDF15 in the circulation can affect metabolic processes. Studies have shown that GDF15 is associated with changes in body weight. SUMMARY This review reviews the current knowledge on the relationship between GDF15 and body weight change, focusing on the role and mechanism of GDF15 in body weight regulation. GDF15 plays an important role in reducing food intake, improving insulin resistance, and breaking down fat, suggesting that GDF15 has an important regulatory effect on body weight. The mechanism by which GDF15 causes reduced food intake may be related to changes in food preference, delayed gastric emptying, and conditioned taste aversion. GDF15 can combat insulin resistance induced by inflammation or protect β cell from apoptosis. GDF15 probably promotes lipolysis through a brain-somatic tissue circuit. Several factors and related signaling pathways are also mentioned that can contribute to the effects of GDF15 on reducing weight. KEY MESSAGE GDF15 plays an important role in weight regulation and provides a new direction for the treatment of obesity. Its effects on resisting obesity are of great significance to inhibiting the progression of metabolic diseases. It is expected to become a new target for regulating body weight, improving obesity, and treating metabolic diseases such as diabetes.
Collapse
Affiliation(s)
- Xiao-Chen Dong
- Department of Internal Cardiovascular Medicine, Second Xiangya Hospital of Central South University, Changsha, China
| | - Dan-Yan Xu
- Department of Internal Cardiovascular Medicine, Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
11
|
Bucurica S, Lupanciuc M, Ionita-Radu F, Stefan I, Munteanu AE, Anghel D, Jinga M, Gaman EL. Estrobolome and Hepatocellular Adenomas-Connecting the Dots of the Gut Microbial β-Glucuronidase Pathway as a Metabolic Link. Int J Mol Sci 2023; 24:16034. [PMID: 38003224 PMCID: PMC10671049 DOI: 10.3390/ijms242216034] [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: 10/08/2023] [Revised: 11/02/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
Hepatocellular adenomas are benign endothelial tumors of the liver, mostly associated with female individual users of estrogen-containing medications. However, the precise factors underlying the selective development of hepatic adenomas in certain females remain elusive. Additionally, the conventional profile of individuals prone to hepatic adenoma is changing. Notably, male patients exhibit a higher risk of malignant progression of hepatocellular adenomas, and there are instances where hepatic adenomas have no identifiable cause. In this paper, we theorize the role of the human gastrointestinal microbiota, specifically, of bacterial species producing β-glucuronidase enzymes, in the development of hepatic adenomas through the estrogen recycling pathway. Furthermore, we aim to address some of the existing gaps in our knowledge of pathophysiological pathways which are not yet subject to research or need to be studied further. As microbial β-glucuronidases proteins recycle estrogen and facilitate the conversion of inactive estrogen into its active form, this process results in elevated levels of unbound plasmatic estrogen, leading to extended exposure to estrogen. We suggest that an imbalance in the estrobolome could contribute to sex hormone disease evolution and, consequently, to the advancement of hepatocellular adenomas, which are estrogen related.
Collapse
Affiliation(s)
- Sandica Bucurica
- Department of Gastroenterology, “Carol Davila” University of Medicine and Pharmacy Bucharest, 020021 Bucharest, Romania;
- Department of Gastroenterology, “Dr. Carol Davila” Central Military Emergency University Hospital, 010242 Bucharest, Romania;
| | - Mihaela Lupanciuc
- Department of Gastroenterology, “Dr. Carol Davila” Central Military Emergency University Hospital, 010242 Bucharest, Romania;
| | - Florentina Ionita-Radu
- Department of Gastroenterology, “Carol Davila” University of Medicine and Pharmacy Bucharest, 020021 Bucharest, Romania;
- Department of Gastroenterology, “Dr. Carol Davila” Central Military Emergency University Hospital, 010242 Bucharest, Romania;
| | - Ion Stefan
- Department of Infectious Diseases, “Dr. Carol Davila” Central Military Emergency University Hospital, 010242 Bucharest, Romania;
- Department of Medico-Surgical and Prophylactic Disciplines, Titu Maiorescu University, 031593 Bucharest, Romania; (A.E.M.); (D.A.)
| | - Alice Elena Munteanu
- Department of Medico-Surgical and Prophylactic Disciplines, Titu Maiorescu University, 031593 Bucharest, Romania; (A.E.M.); (D.A.)
- Department of Cardiology, “Dr. Carol Davila” Central Military Emergency University Hospital, 010242 Bucharest, Romania
| | - Daniela Anghel
- Department of Medico-Surgical and Prophylactic Disciplines, Titu Maiorescu University, 031593 Bucharest, Romania; (A.E.M.); (D.A.)
- Department of Internal Medicine, “Dr. Carol Davila” Central Military Emergency University Hospital, 010242 Bucharest, Romania
| | - Mariana Jinga
- Department of Gastroenterology, “Carol Davila” University of Medicine and Pharmacy Bucharest, 020021 Bucharest, Romania;
- Department of Gastroenterology, “Dr. Carol Davila” Central Military Emergency University Hospital, 010242 Bucharest, Romania;
| | - Elena Laura Gaman
- Department of Biochemistry, “Carol Davila” University of Medicine and Pharmacy Bucharest, 020021 Bucharest, Romania;
| |
Collapse
|
12
|
Oliviero F, Klement W, Mary L, Dauwe Y, Lippi Y, Naylies C, Gayrard V, Marchi N, Mselli-Lakhal L. CAR Protects Females from Diet-Induced Steatosis and Associated Metabolic Disorders. Cells 2023; 12:2218. [PMID: 37759441 PMCID: PMC10527310 DOI: 10.3390/cells12182218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 08/30/2023] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
Non-Alcoholic Fatty Liver Disease (NAFLD) is the most common cause of chronic liver disease worldwide, affecting 70-90% of obese individuals. In humans, a lower NAFLD incidence is reported in pre-menopausal women, although the mechanisms affording this protection remain under-investigated. Here, we tested the hypothesis that the constitutive androstane nuclear receptor (CAR) plays a role in the pathogenesis of experimental NAFLD. Male and female wild-type (WT) and CAR knock-out (CAR-/-) mice were subjected to a high-fat diet (HFD) for 16 weeks. We examined the metabolic phenotype of mice through body weight follow-up, glucose tolerance tests, analysis of plasmatic metabolic markers, hepatic lipid accumulation, and hepatic transcriptome. Finally, we examined the potential impact of HFD and CAR deletion on specific brain regions, focusing on glial cells. HFD-induced weight gain and hepatic steatosis are more pronounced in WT males than females. CAR-/- females present a NASH-like hepatic transcriptomic signature suggesting a potential NAFLD to NASH transition. Transcriptomic correlation analysis highlighted a possible cross-talk between CAR and ERα receptors. The peripheral effects of CAR deletion in female mice were associated with astrogliosis in the hypothalamus. These findings prove that nuclear receptor CAR may be a potential mechanism entry-point and a therapeutic target for treating NAFLD/NASH.
Collapse
Affiliation(s)
- Fabiana Oliviero
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31027 Toulouse, France
| | - Wendy Klement
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, 34094 Montpellier, France
| | - Lucile Mary
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31027 Toulouse, France
| | - Yannick Dauwe
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31027 Toulouse, France
| | - Yannick Lippi
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31027 Toulouse, France
| | - Claire Naylies
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31027 Toulouse, France
| | - Véronique Gayrard
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31027 Toulouse, France
| | - Nicola Marchi
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31027 Toulouse, France
| | - Laila Mselli-Lakhal
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31027 Toulouse, France
| |
Collapse
|
13
|
Kasarinaite A, Sinton M, Saunders PTK, Hay DC. The Influence of Sex Hormones in Liver Function and Disease. Cells 2023; 12:1604. [PMID: 37371074 PMCID: PMC10296738 DOI: 10.3390/cells12121604] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/09/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
The liver performs a multitude of bodily functions, whilst retaining the ability to regenerate damaged tissue. In this review, we discuss sex steroid biology, regulation of mammalian liver physiology and the development of new model systems to improve our understanding of liver biology in health and disease. A major risk factor for the development of liver disease is hepatic fibrosis. Key drivers of this process are metabolic dysfunction and pathologic activation of the immune system. Although non-alcoholic fatty liver disease (NAFLD) is largely regarded as benign, it does progress to non-alcoholic steatohepatitis in a subset of patients, increasing their risk of developing cirrhosis and hepatocellular carcinoma. NAFLD susceptibility varies across the population, with obesity and insulin resistance playing a strong role in the disease development. Additionally, sex and age have been identified as important risk factors. In addition to the regulation of liver biochemistry, sex hormones also regulate the immune system, with sexual dimorphism described for both innate and adaptive immune responses. Therefore, sex differences in liver metabolism, immunity and their interplay are important factors to consider when designing, studying and developing therapeutic strategies to treat human liver disease. The purpose of this review is to provide the reader with a general overview of sex steroid biology and their regulation of mammalian liver physiology.
Collapse
Affiliation(s)
- Alvile Kasarinaite
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, Edinburgh EH16 4UU, UK
| | - Matthew Sinton
- School of Biodiversity, One Health, and Veterinary Medicine, University of Glasgow, Glasgow G61 1QH, UK
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow G12 9TA, UK
| | - Philippa T. K. Saunders
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, Edinburgh EH16 4UU, UK
| | - David C. Hay
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, Edinburgh EH16 4UU, UK
| |
Collapse
|
14
|
Isola JVV, Ko S, Ocañas SR, Stout MB. Role of Estrogen Receptor α in Aging and Chronic Disease. ADVANCES IN GERIATRIC MEDICINE AND RESEARCH 2023; 5:e230005. [PMID: 37425648 PMCID: PMC10327608 DOI: 10.20900/agmr20230005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Estrogen receptor alpha (ERα) plays a crucial role in reproductive function in both sexes. It also mediates cellular responses to estrogens in multiple nonreproductive organ systems, many of which regulate systemic metabolic homeostasis and inflammatory processes in mammals. The loss of estrogens and/or ERα agonism during aging is associated with the emergence of several comorbid conditions, particularly in females undergoing the menopausal transition. Emerging data also suggests that male mammals likely benefit from ERα agonism if done in a way that circumvents feminizing characteristics. This has led us, and others, to speculate that tissue-specific ERα agonism may hold therapeutic potential for curtailing aging and chronic disease burden in males and females that are at high-risk of cancer and/or cardiovascular events with traditional estrogen replacement therapies. In this mini-review, we emphasize the role of ERα in the brain and liver, summarizing recent evidence that indicates these two organs systems mediate the beneficial effects of estrogens on metabolism and inflammation during aging. We also discuss how 17α-estradiol administration elicits health benefits in an ERα-dependent manner, which provides proof-of-concept that ERα may be a druggable target for attenuating aging and age-related disease burden.
Collapse
Affiliation(s)
- José V. V. Isola
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Sunghwan Ko
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Sarah R. Ocañas
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA
| | - Michael B. Stout
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
15
|
Fuller KNZ, Allen J, Kumari R, Akakpo JY, Ruebel M, Shankar K, Thyfault JP. Pre- and Post-Sexual Maturity Liver-specific ERα Knockout Does Not Impact Hepatic Mitochondrial Function. J Endocr Soc 2023; 7:bvad053. [PMID: 37197409 PMCID: PMC10184454 DOI: 10.1210/jendso/bvad053] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Indexed: 05/19/2023] Open
Abstract
Compared with males, premenopausal women and female rodents are protected against hepatic steatosis and present with higher functioning mitochondria (greater hepatic mitochondrial respiration and reduced H2O2 emission). Despite evidence that estrogen action mediates female protection against steatosis, mechanisms remain unknown. Here we validated a mouse model with inducible reduction of liver estrogen receptor alpha (ERα) (LERKO) via adeno-associated virus (AAV) Cre. We phenotyped the liver health and mitochondrial function of LERKO mice (n = 10-12 per group) on a short-term high-fat diet (HFD), and then tested whether timing of LERKO induction at 2 timepoints (sexually immature: 4 weeks old [n = 11 per group] vs sexually mature: 8-10 weeks old [n = 8 per group]) would impact HFD-induced outcomes. We opted for an inducible LERKO model due to known estrogen-mediated developmental programming, and we reported both receptor and tissue specificity with our model. Control mice were ERαfl/fl receiving AAV with green fluorescent protein (GFP) only. Results show that there were no differences in body weight/composition or hepatic steatosis in LERKO mice with either short-term (4-week) or chronic (8-week) high-fat feeding. Similarly, LERKO genotype nor timing of LERKO induction (pre vs post sexual maturity) did not alter hepatic mitochondrial O2 and H2O2 flux, coupling, or OXPHOS protein. Transcriptomic analysis showed that hepatic gene expression in LERKO was significantly influenced by developmental stage. Together, these studies suggest that hepatic ERα is not required in female protection against HFD-induced hepatic steatosis nor does it mediate sexual dimorphism in liver mitochondria function.
Collapse
Affiliation(s)
- Kelly N Z Fuller
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, KS 64128, USA
| | - Julie Allen
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, KS 64128, USA
| | - Roshan Kumari
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, KS 64128, USA
| | - Jephte Y Akakpo
- Department of Pharmacology and Toxicology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Meghan Ruebel
- USDA-ARS, Southeast Area, Arkansas Children’s Nutrition Center, Little Rock, AR 72202, USA
| | - Kartik Shankar
- USDA-ARS, Southeast Area, Arkansas Children’s Nutrition Center, Little Rock, AR 72202, USA
| | - John P Thyfault
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, KS 64128, USA
- KU Diabetes Institute and Kansas Center for Metabolism and Obesity, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Internal Medicine, Division of Endocrinology and Metabolism, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Center for Children’s Healthy Lifestyles and Nutrition, Kansas City, MO 64108, USA
| |
Collapse
|
16
|
Fang Z, Xu H, Duan J, Ruan B, Liu J, Song P, Ding J, Xu C, Li Z, Dou K, Wang L. Short-term tamoxifen administration improves hepatic steatosis and glucose intolerance through JNK/MAPK in mice. Signal Transduct Target Ther 2023; 8:94. [PMID: 36864030 PMCID: PMC9981902 DOI: 10.1038/s41392-022-01299-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 11/30/2022] [Accepted: 12/22/2022] [Indexed: 03/04/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) which is a leading cause of chronic liver diseases lacks effective treatment. Tamoxifen has been proven to be the first-line chemotherapy for several solid tumors in clinics, however, its therapeutic role in NAFLD has never been elucidated before. In vitro experiments, tamoxifen protected hepatocytes against sodium palmitate-induced lipotoxicity. In male and female mice fed with normal diets, continuous tamoxifen administration inhibited lipid accumulation in liver, and improved glucose and insulin intolerance. Short-term tamoxifen administration largely improved hepatic steatosis and insulin resistance, however, the phenotypes manifesting inflammation and fibrosis remained unchanged in abovementioned models. In addition, mRNA expressions of genes related to lipogenesis, inflammation, and fibrosis were downregulated by tamoxifen treatment. Moreover, the therapeutic effect of tamoxifen on NAFLD was not gender or ER dependent, as male and female mice with metabolic disorders shared no difference in response to tamoxifen and ER antagonist (fulvestrant) did not abolish its therapeutic effect as well. Mechanistically, RNA sequence of hepatocytes isolated from fatty liver revealed that JNK/MAPK signaling pathway was inactivated by tamoxifen. Pharmacological JNK activator (anisomycin) partially deprived the therapeutic role of tamoxifen in treating hepatic steatosis, proving tamoxifen improved NAFLD in a JNK/MAPK signaling-dependent manner.
Collapse
Affiliation(s)
- Zhiqiang Fang
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Hao Xu
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Juanli Duan
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Bai Ruan
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China.,Center of Clinical Aerospace Medicine & Department of Aviation Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Jingjing Liu
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Ping Song
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jian Ding
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Chen Xu
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Zhiwen Li
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Kefeng Dou
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Lin Wang
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
17
|
Fuller KNZ, McCoin CS, Stierwalt H, Allen J, Gandhi S, Perry CGR, Jambal P, Shankar K, Thyfault JP. Oral combined contraceptives induce liver mitochondrial reactive oxygen species and whole-body metabolic adaptations in female mice. J Physiol 2022; 600:5215-5245. [PMID: 36326014 DOI: 10.1113/jp283733] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022] Open
Abstract
Compared to age-matched men, pre-menopausal women show greater resilience against cardiovascular disease (CVD), hepatic steatosis, diabetes and obesity - findings that are widely attributed to oestrogen. However, meta-analysis data suggest that current use of oral combined contraceptives (OC) is a risk factor for myocardial infarction, and OC use further compounds with metabolic disease risk factors to increase CVD susceptibility. While mitochondrial function in tissues such as the liver and skeletal muscle is an emerging mechanism by which oestrogen may confer its protection, effects of OC use on mitochondria and metabolism in the context of disease risk remain unexplored. To answer this question, female C57Bl/6J mice were fed a high fat diet and treated with vehicle or OCs for 3, 12 or 20 weeks (n = 6 to 12 per group) at a dose and ratio that mimic the human condition of cycle cessation in the low oestrogen, high progesterone stage. Liver and skeletal muscle mitochondrial function (respiratory capacity, H2 O2 , coupling) was measured along with clinical outcomes of cardiometabolic disease such as obesity, glucose tolerance, hepatic steatosis and aortic atherosclerosis. The main findings indicate that regardless of treatment duration, OCs robustly increase hepatic mitochondrial H2 O2 levels, likely due to diminished antioxidant capacity, but have no impact on muscle mitochondrial H2 O2 . Furthermore, OC-treated mice had lower adiposity and hepatic triglyceride content compared to control mice despite reduced wheel running, spontaneous physical activity and total energy expenditure. Together, these studies describe tissue-specific effects of OC use on mitochondria as well as variable impacts on markers of metabolic disease susceptibility. KEY POINTS: Oestrogen loss in women increases risk for cardiometabolic diseases, a link that has been partially attributed to negative impacts on mitochondria and energy metabolism. To study the effect of oral combined contraceptives (OCs) on hepatic and skeletal muscle mitochondria and whole-body energy metabolism, we used an animal model of OCs which mimics the human condition of cessation of hormonal cycling in the low oestrogen, high progesterone state. OC-treated mice have increased hepatic mitochondrial oxidative stress and decreased physical activity and energy expenditure, despite displaying lower adiposity and liver fat at this time point. These pre-clinical data reveal tissue-specific effects of OCs that likely underlie the clinical findings of increased cardiometabolic disease in women who use OCs compared to non-users, when matched for obesity.
Collapse
Affiliation(s)
- Kelly N Z Fuller
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA.,Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, MO, USA
| | - Colin S McCoin
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA.,Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, MO, USA.,Center for Children's Healthy Lifestyles and Nutrition, Kansas City, MO, USA.,University of Kansas Diabetes Institute, Kansas City, KS, USA.,Kansas Center for Metabolism and Obesity Research, Kansas City, KS, USA
| | - Harrison Stierwalt
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA.,Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, MO, USA
| | - Julie Allen
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA.,Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, MO, USA
| | - Shivam Gandhi
- School of Kinesiology and Health Science, Muscle Health Research Center, York University, Toronto, Canada
| | - Christopher G R Perry
- School of Kinesiology and Health Science, Muscle Health Research Center, York University, Toronto, Canada
| | - Purevsuren Jambal
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine Anschutz Medical Campus, Aurora, CO, USA
| | - Kartik Shankar
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine Anschutz Medical Campus, Aurora, CO, USA
| | - John P Thyfault
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA.,Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, MO, USA.,Center for Children's Healthy Lifestyles and Nutrition, Kansas City, MO, USA.,University of Kansas Diabetes Institute, Kansas City, KS, USA.,Kansas Center for Metabolism and Obesity Research, Kansas City, KS, USA.,Department of Internal Medicine, Division of Endocrinology and Metabolism, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
18
|
Wang Y, Chen J, Chen C, Peng H, Lin X, Zhao Q, Chen S, Wang X. Growth differentiation factor-15 overexpression promotes cell proliferation and predicts poor prognosis in cerebral lower-grade gliomas correlated with hypoxia and glycolysis signature. Life Sci 2022; 302:120645. [PMID: 35588865 DOI: 10.1016/j.lfs.2022.120645] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 04/15/2022] [Accepted: 05/12/2022] [Indexed: 12/30/2022]
Abstract
AIMS Growth differentiation factor-15 (GDF15) plays complex and controversial roles in cancer. In this study, the prognostic value and the exact biological function of GDF15 in cerebral lower-grade gliomas (LGGs) and its potential molecular targets were examined. MAIN METHODS Wilcoxon signed-rank test and logistic regression were applied to analyze associations between GDF15 expression and clinical characteristics using the Cancer Genome Atlas (TCGA) database. Overall survival was analyzed using Kaplan-Meier and Cox analyses. Gene set enrichment analysis (GSEA) and the hypoxia risk model was conducted to identify the potential molecular mechanisms underlying the effects of GDF15 on LGGs tumorigenesis. The biological function of GDF15 was examined using gain- and loss-of-function experiments, and a recombinant hGDF15 protein in LGG SW1783 cells in vitro. KEY FINDINGS We found that higher GDF15 expression is associated with poor clinical features in LGG patients, and an independent risk factor for overall survival among LGG patients. GSEA results showed that the poor prognostic role of GDF15 in LGGs is related to hypoxia and glycolysis signatures, which was further validated using the hypoxia risk model. Furthermore, GDF15 overexpression facilitated cell proliferation, while GDF15 siRNA inhibits cell proliferation in LGG SW1783 cells. In addition, GDF15 was upregulated upon CoCl2 treatment which induces hypoxia, correlating with the upregulation of the expressions of HIF-1α and glycolysis-related key genes in SW1783 cells. SIGNIFICANCE GDF15 may promote LGG tumorigenesis that is associated with the hypoxia and glycolysis pathways, and thus could serve as a promising molecular target for LGG prevention and therapy.
Collapse
Affiliation(s)
- Ying Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Jiajun Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Chaojie Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - He Peng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Xiaojian Lin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Qian Zhao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Shengjia Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Xingya Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, PR China.
| |
Collapse
|
19
|
Wang Y, Chen C, Chen J, Sang T, Peng H, Lin X, Zhao Q, Chen S, Eling T, Wang X. Overexpression of NAG-1/GDF15 prevents hepatic steatosis through inhibiting oxidative stress-mediated dsDNA release and AIM2 inflammasome activation. Redox Biol 2022; 52:102322. [PMID: 35504134 PMCID: PMC9079118 DOI: 10.1016/j.redox.2022.102322] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/10/2022] [Accepted: 04/23/2022] [Indexed: 02/08/2023] Open
Abstract
Mitochondrial dysfunction and oxidative stress-mediated inflammasome activation play critical roles in the pathogenesis of the non-alcoholic fatty liver disease (NAFLD). Non-steroidal anti-inflammatory drug (NSAID)-activated gene-1 (NAG-1), or growth differentiation factor-15 (GDF15), is associated with many biological processes and diseases, including NAFLD. However, the role of NAG-1/GDF15 in regulating oxidative stress and whether this process is associated with absent in melanoma 2 (AIM2) inflammasome activation in NAFLD are unknown. In this study, we revealed that NAG-1/GDF15 is significantly downregulated in liver tissues of patients with steatosis compared to normal livers using the Gene Expression Omnibus (GEO) database, and in free fatty acids (FFA, oleic acid/palmitic acid, 2:1)-induced HepG2 and Huh-7 cellular steatosis models. Overexpression of NAG-1/GDF15 in transgenic (Tg) mice significantly alleviated HFD-induced obesity and hepatic steatosis, improved lipid homeostasis, enhanced fatty acid β-oxidation and lipolysis, inhibited fatty acid synthesis and uptake, and inhibited AIM2 inflammasome activation and the secretion of IL-18 and IL-1β, as compared to their wild-type (WT) littermates without reducing food intake. Furthermore, NAG-1/GDF15 overexpression attenuated FFA-induced triglyceride (TG) accumulation, lipid metabolism deregulation, and AIM2 inflammasome activation in hepatic steatotic cells, while knockdown of NAG-1/GDF15 demonstrated opposite effects. Moreover, NAG-1/GDF15 overexpression inhibited HFD- and FFA-induced oxidative stress and mitochondrial damage which in turn reduced double-strand DNA (dsDNA) release into the cytosol, while NAG-1/GDF15 siRNA showed opposite effects. The reduced ROS production and dsDNA release may be responsible for attenuated AIM2 activation by NAG-1/GDF15 upon fatty acid overload. In conclusion, our results provide evidence that other than regulating lipid homeostasis, NAG-1/GDF15 protects against hepatic steatosis through a novel mechanism via suppressing oxidative stress, mitochondrial damage, dsDNA release, and AIM2 inflammasome activation. NAG-1/GDF15 is downregulated in human steatotic liver and FFA-induced liver cells. NAG-1/GDF15 inhibits hepatic steatosis and improves lipid homeostasis. AIM2 inflammasome is activated in steatosis models and is inhibited by NAG-1/GDF15. NAG-1/GDF15 reduces oxidative stress and mitochondrial damage in steatosis models. NAG-1/GDF15 inhibits mitochondrial dsDNA release and thus inhibits AIM2 activation.
Collapse
|
20
|
Stokar J, Gurt I, Cohen-Kfir E, Yakubovsky O, Hallak N, Benyamini H, Lishinsky N, Offir N, Tam J, Dresner-Pollak R. Hepatic Adropin is Regulated by Estrogen and Contributes to Adverse Metabolic Phenotypes in Ovariectomized Mice. Mol Metab 2022; 60:101482. [PMID: 35364299 PMCID: PMC9044006 DOI: 10.1016/j.molmet.2022.101482] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/13/2022] [Accepted: 03/24/2022] [Indexed: 12/02/2022] Open
Abstract
Objective Menopause is associated with visceral adiposity, hepatic steatosis and increased risk for cardiovascular disease. As estrogen replacement therapy is not suitable for all postmenopausal women, a need for alternative therapeutics and biomarkers has emerged. Methods 9-week-old C57BL/6 J female mice were subjected to ovariectomy (OVX) or SHAM surgery (n = 10 per group), fed a standard diet and sacrificed 6- & 12 weeks post-surgery. Results Increased weight gain, hepatic triglyceride content and changes in hepatic gene expression of Cyp17a1, Rgs16, Fitm1 as well as Il18, Rares2, Retn, Rbp4 in mesenteric visceral adipose tissue (VAT) were observed in OVX vs. SHAM. Liver RNA-sequencing 6-weeks post-surgery revealed changes in genes and microRNAs involved in fat metabolism in OVX vs. SHAM mice. Energy Homeostasis Associated gene (Enho) coding for the hepatokine adropin was significantly reduced in OVX mice livers and strongly inversely correlated with weight gain (r = −0.7 p < 0.001) and liver triglyceride content (r = −0.4, p = 0.04), with a similar trend for serum adropin. In vitro, Enho expression was tripled by 17β-estradiol in BNL 1 ME liver cells with increased adropin in supernatant. Analysis of open-access datasets revealed increased hepatic Enho expression in estrogen treated OVX mice and estrogen dependent ERα binding to Enho. Treatment of 5-month-old OVX mice with Adropin (i.p. 450 nmol/kg/twice daily, n = 4,5 per group) for 6-weeks reversed adverse adipokine gene expression signature in VAT, with a trended increase in lean body mass and decreased liver TG content with upregulation of Rgs16. Conclusions OVX is sufficient to induce deranged metabolism in adult female mice. Hepatic adropin is regulated by estrogen, negatively correlated with adverse OVX-induced metabolic phenotypes, which were partially reversed with adropin treatment. Adropin should be further explored as a potential therapeutic target and biomarker for menopause-related metabolic derangement. OVX increased body weight, liver fat & adverse visceral fat adipokine signature. OVX altered liver transcriptome & miRNA profile including fat metabolism pathways. Enho was downregulated by OVX & inversely correlated with weight gain & liver fat. Hepatic adropin expression was upregulated by estrogen in-vitro & in-vivo. Adropin treatment partially reversed OVX induced adverse metabolic phenotypes.
Collapse
|
21
|
Yi M, Shindo S, Negishi M. Immunoprecipitation Analyses of Estrogen Receptor α Phosphorylated at Serine 216 in the Mouse Liver. Methods Mol Biol 2022; 2418:41-51. [PMID: 35119658 DOI: 10.1007/978-1-0716-1920-9_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Estrogen receptor α (ERα) conserves a phosphorylation motif at Serine 216. This site constitutes a protein kinase C phosphorylation motif located within the DNA binding domain (DBD) of ERα. The liver plays a critical role in the regulation of metabolism of various xenobiotics, fatty acids, and cholesterol or endogenous compounds. Moreover, numerous metabolizing enzymes are mainly expressed in the liver. In this chapter, we describe several practical experimental procedures confirming that mouse ERα is phosphorylated at serine 216 in livers upon phenobarbital (PB) treatment. Also, this phosphorylation regulates the expression of estrogen sulfotransferase gene (SULT1E1) which has an important role to sulfate and inactivate estrogen. In response to PB, the conserved motif within the DBD activates the Sult1e1 gene. When this motif was mutated, the activation of Sult1e1 was suppressed significantly. This chapter also describes the use of a phospho-peptide antibody (αP-S216) in the chromatin immunoprecipitation (ChIP) assay, and the co-immunoprecipitation (Co-IP) assay visualized by Western blot analysis.
Collapse
Affiliation(s)
- MyeongJin Yi
- Pharmacogenetics Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC, USA.
| | - Sawako Shindo
- Pharmacogenetics Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC, USA
- Department of Environmental Health, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Masahiko Negishi
- Pharmacogenetics Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC, USA
| |
Collapse
|
22
|
Huang R, Guo F, Li Y, Liang Y, Li G, Fu P, Ma L. Activation of AMPK by triptolide alleviates nonalcoholic fatty liver disease by improving hepatic lipid metabolism, inflammation and fibrosis. PHYTOMEDICINE 2021; 92:153739. [PMID: 34592488 DOI: 10.1016/j.phymed.2021.153739] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 07/29/2021] [Accepted: 09/05/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Triptolide is naturally isolated from Tripterygium wilfordii Hook F., possessing multiple biological activities. Hepatotoxicity is one of the main side effects of triptolide. However, the effect of triptolide on nonalcoholic fatty liver disease remains unknown (NAFLD). PURPOSE This study aimed to observe the amelioration of triptolide against NAFLD and investigate the engaged mechanism. METHODS Two typical animal models of NAFLD, obese db/db mice and methionine/choline-deficient (MCD) diet-fed mice, were used. Hepatic steatosis, inflammation, and fibrosis were evaluated by H&E and Masson staining. Oil red O staining and lipid extraction analysis were used to detect fat content in mice livers. Expression of lipid metabolism, inflammatory and fibrogenic genes was also detected by Real-time PCR and Western blotting, respectively. Phosphoproteomics, molecular docking, and TR-FRET assay were performed to provide further insight into how triptolide improved NAFLD. RESULTS Intraperitoneal injection of triptolide at a daily dose of 50 μg/kg significantly alleviated MCD diet-induced nonalcoholic steatohepatitis (NASH), but 100 μg/kg triptolide caused severe hepatotoxicity. Pathological staining confirmed low-dose triptolide treatment reducing hepatic lipid deposition, inflammation, and fibrosis in NASH. Serum biochemical analysis revealed a reduction in the level of liver enzymes and bilirubin. MCD also induced rising expression of typical genes and proteins related to fibrosis (fibronectin, α-SMA, collagens, TGF-β) and inflammation (ILs, TNF-α, MCP-1), which was suppressed by low-dose triptolide. Data from the proteomics/phosphoproteomics and TR-FRET assay indicated triptolide was a potential allosteric AMPK agonist to increase the phosphorylation on Thr172 residue, with the EC50 of 277.78 μM and 231.02 μM for AMPKα1 and AMPKα2, respectively. Moreover, triptolide exhibited an ability to activate AMPK and further led to increasing ACC1 phosphorylation in the liver. The positive results that triptolide ameliorated hepatic lipogenesis, fatty acid oxidation, and fibrosis of NAFLD via activating AMPK were further confirmed in db/db mice with 10-week intervention (50 μg/kg, i.v., twice a week). CONCLUSION This study demonstrates that dose-related triptolide as an allosteric AMPK agonist has the potential to alleviate NAFLD without hepatotoxicity.
Collapse
Affiliation(s)
- Rongshuang Huang
- Division of Nephrology and National Clinical Research Center for Geriatrics, Kidney Research Institute, West China Hospital of Sichuan University, No.37 Guoxue Alley, Chengdu 610041, China
| | - Fan Guo
- Division of Nephrology and National Clinical Research Center for Geriatrics, Kidney Research Institute, West China Hospital of Sichuan University, No.37 Guoxue Alley, Chengdu 610041, China
| | - Yanping Li
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Yan Liang
- Research Core Facility of West China Hospital, Sichuan University, Chengdu 610041, China
| | - Guobo Li
- West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Ping Fu
- Division of Nephrology and National Clinical Research Center for Geriatrics, Kidney Research Institute, West China Hospital of Sichuan University, No.37 Guoxue Alley, Chengdu 610041, China.
| | - Liang Ma
- Division of Nephrology and National Clinical Research Center for Geriatrics, Kidney Research Institute, West China Hospital of Sichuan University, No.37 Guoxue Alley, Chengdu 610041, China.
| |
Collapse
|
23
|
Della Torre S. Beyond the X Factor: Relevance of Sex Hormones in NAFLD Pathophysiology. Cells 2021; 10:2502. [PMID: 34572151 PMCID: PMC8470830 DOI: 10.3390/cells10092502] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/12/2021] [Accepted: 09/14/2021] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a major health issue worldwide, being frequently associated with obesity, unbalanced dietary regimens, and reduced physical activity. Despite their greater adiposity and reduced physical activity, women show a lower risk of developing NAFLD in comparison to men, likely a consequence of a sex-specific regulation of liver metabolism. In the liver, sex differences in the uptake, synthesis, oxidation, deposition, and mobilization of lipids, as well as in the regulation of inflammation, are associated with differences in NAFLD prevalence and progression between men and women. Given the major role of sex hormones in driving hepatic sexual dimorphism, this review will focus on the role of sex hormones and their signaling in the regulation of hepatic metabolism and in the molecular mechanisms triggering NAFLD development and progression.
Collapse
Affiliation(s)
- Sara Della Torre
- Department of Pharmaceutical Sciences, University of Milan, Via Balzaretti 9, 20133 Milan, Italy
| |
Collapse
|
24
|
Morán-Costoya A, Proenza AM, Gianotti M, Lladó I, Valle A. Sex Differences in Nonalcoholic Fatty Liver Disease: Estrogen Influence on the Liver-Adipose Tissue Crosstalk. Antioxid Redox Signal 2021; 35:753-774. [PMID: 33736456 DOI: 10.1089/ars.2021.0044] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Significance: Nonalcoholic fatty liver disease (NAFLD) is a hepatic and systemic disorder with a complex multifactorial pathogenesis. Owing to the rising incidence of obesity and diabetes mellitus, the prevalence of NAFLD and its impact on global health care are expected to increase in the future. Differences in NAFLD exist between males and females, and among females depending on their reproductive status. Clinical and preclinical data show that females in the fertile age are more protected against NAFLD, and studies in postmenopausal women and ovariectomized animal models support a protective role for estrogens. Recent Advances: An efficient crosstalk between the liver and adipose tissue is necessary to regulate lipid and glucose metabolism, protecting the liver from steatosis and insulin resistance contributing to NALFD. New advances in the knowledge of sexual dimorphism in liver and adipose tissue are providing interesting clues about the sex differences in NAFLD pathogenesis that could inspire new therapeutic strategies. Critical Issues: Sex hormones influence key master regulators of lipid metabolism and oxidative stress in liver and adipose tissue. All these sex-biased metabolic adjustments shape the crosstalk between liver and adipose tissue, contributing to the higher protection of females to NAFLD. Future Directions: The development of novel drugs based on the protective action of estrogens, but without its feminizing or undesired side effects, might provide new therapeutic strategies for the management of NAFLD. Antioxid. Redox Signal. 35, 753-774.
Collapse
Affiliation(s)
- Andrea Morán-Costoya
- Energy Metabolism and Nutrition Group, Department of Fundamental Biology and Health Sciences, Research Institute of Health Sciences (IUNICS), University of the Balearic Islands, Palma, Spain.,Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Ana M Proenza
- Energy Metabolism and Nutrition Group, Department of Fundamental Biology and Health Sciences, Research Institute of Health Sciences (IUNICS), University of the Balearic Islands, Palma, Spain.,Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain.,Center for Biomedical Research in the Pathophysiology of Obesity and Nutrition Network, Carlos III Health Institute, Madrid, Spain
| | - Magdalena Gianotti
- Energy Metabolism and Nutrition Group, Department of Fundamental Biology and Health Sciences, Research Institute of Health Sciences (IUNICS), University of the Balearic Islands, Palma, Spain.,Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain.,Center for Biomedical Research in the Pathophysiology of Obesity and Nutrition Network, Carlos III Health Institute, Madrid, Spain
| | - Isabel Lladó
- Energy Metabolism and Nutrition Group, Department of Fundamental Biology and Health Sciences, Research Institute of Health Sciences (IUNICS), University of the Balearic Islands, Palma, Spain.,Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain.,Center for Biomedical Research in the Pathophysiology of Obesity and Nutrition Network, Carlos III Health Institute, Madrid, Spain
| | - Adamo Valle
- Energy Metabolism and Nutrition Group, Department of Fundamental Biology and Health Sciences, Research Institute of Health Sciences (IUNICS), University of the Balearic Islands, Palma, Spain.,Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain.,Center for Biomedical Research in the Pathophysiology of Obesity and Nutrition Network, Carlos III Health Institute, Madrid, Spain
| |
Collapse
|
25
|
Estrada-Meza J, Videlo J, Bron C, Saint-Béat C, Silva M, Duboeuf F, Peyruchaud O, Rajas F, Mithieux G, Gautier-Stein A. Tamoxifen Treatment in the Neonatal Period Affects Glucose Homeostasis in Adult Mice in a Sex-Dependent Manner. Endocrinology 2021; 162:6277101. [PMID: 33999998 DOI: 10.1210/endocr/bqab098] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Indexed: 12/11/2022]
Abstract
Tamoxifen is a selective estrogen receptor modulator used to activate the CREERT2 recombinase, allowing tissue-specific and temporal control of the somatic mutagenesis to generate transgenic mice. Studies integrating development and metabolism require a genetic modification induced by a neonatal tamoxifen administration. Here, we investigate the effects of a neonatal tamoxifen administration on energy homeostasis in adult male and female C57BL/6J mice. C57BL/6J male and female mouse pups received a single injection of tamoxifen 1 day after birth (NTT) and were fed a high-fat/high-sucrose diet at 6 weeks of age. We measured weight, body composition, glucose and insulin tolerance, basal metabolism, and tibia length and weight in adult mice. The neonatal tamoxifen administration exerted long-term, sex-dependent effects on energy homeostasis. NTT female mice became overweight and developed impaired glucose control in comparison to vehicle-treated littermates. NTT females exhibited 60% increased fat mass, increased food intake, decreased physical activity and energy expenditure, impaired glucose and insulin tolerance, and fasting hyperglycemia and hyperinsulinemia. In contrast, NTT male mice exhibited a modest amelioration of glucose and insulin tolerance and long-term decreased lean mass linked to decreased bone weight. These results suggest that the neonatal tamoxifen administration exerted a marked and sex-dependent influence on adult energy homeostasis and bone weight and must therefore be used with caution for the development of transgenic mouse models regarding studies on energy homeostasis and bone biology.
Collapse
Affiliation(s)
- Judith Estrada-Meza
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - Jasmine Videlo
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - Clara Bron
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - Cécile Saint-Béat
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - Marine Silva
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - François Duboeuf
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1033, Lyon, France
| | - Olivier Peyruchaud
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1033, Lyon, France
| | - Fabienne Rajas
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - Gilles Mithieux
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | | |
Collapse
|
26
|
Guillemot-Legris O, Muccioli GG. The oxysterome and its receptors as pharmacological targets in inflammatory diseases. Br J Pharmacol 2021; 179:4917-4940. [PMID: 33817775 DOI: 10.1111/bph.15479] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 03/14/2021] [Accepted: 03/17/2021] [Indexed: 12/15/2022] Open
Abstract
Oxysterols have gained attention over the last decades and are now considered as fully fledged bioactive lipids. The study of their levels in several conditions, including atherosclerosis, obesity and neurodegenerative diseases, led to a better understanding of their involvement in (patho)physiological processes such as inflammation and immunity. For instance, the characterization of the cholesterol-7α,25-dihydroxycholesterol/GPR183 axis and its implication in immunity represents an important step in the oxysterome study. Besides this axis, others were identified as important in several inflammatory pathologies (such as colitis, lung inflammation and atherosclerosis). However, the oxysterome is a complex system notably due to a redundancy of metabolic enzymes and a wide range of receptors. Indeed, deciphering oxysterol roles and identifying the potential receptor(s) involved in a given pathology remain challenging. Oxysterol properties are very diverse, but most of them could be connected by a common component: inflammation. Here, we review the implication of oxysterol receptors in inflammatory diseases.
Collapse
Affiliation(s)
- Owein Guillemot-Legris
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Giulio G Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
27
|
Le Magueresse-Battistoni B. Endocrine disrupting chemicals and metabolic disorders in the liver: What if we also looked at the female side? CHEMOSPHERE 2021; 268:129212. [PMID: 33359838 DOI: 10.1016/j.chemosphere.2020.129212] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/01/2020] [Accepted: 12/02/2020] [Indexed: 05/07/2023]
Abstract
Endocrine disrupting chemicals (EDCs) are linked to the worldwide epidemic incidence of metabolic disorders and fatty liver diseases, which affects quality of life and represents a high economic cost to society. Energy homeostasis exhibits strong sexual dimorphic traits, and metabolic organs respond to EDCs depending on sex, such as the liver, which orchestrates both drug elimination and glucose and lipid metabolism. In addition, fatty liver diseases show a strong sexual bias, which in part could also originate from sex differences observed in gut microbiota. The aim of this review is to highlight significant differences in endocrine and metabolic aspects of the liver, between males and females throughout development and into adulthood. It is also to illustrate how the male and female liver differently cope with exposure to various EDCs such as bisphenols, phthalates and persistent organic chemicals in order to draw attention to the need to include both sexes in experimental studies. Interesting data come from analyses of the composition and diversity of the gut microbiota in males exposed to the mentioned EDCs showing significant correlations with hepatic lipid accumulation and metabolic disorders but information on females is lacking or incomplete. As industrialization increases, the list of anthropogenic chemicals to which humans will be exposed will also likely increase. In addition to strengthening existing regulations, encouraging populations to protect themselves and promoting the substitution of harmful chemicals with safe products, innovative strategies based on sex differences in the gut microbiota and in the gut-liver axis could be optimistic outlook.
Collapse
|
28
|
Buscato M, Davezac M, Zahreddine R, Adlanmerini M, Métivier R, Fillet M, Cobraiville G, Moro C, Foidart JM, Lenfant F, Gourdy P, Arnal JF, Fontaine C. Estetrol prevents Western diet-induced obesity and atheroma independently of hepatic estrogen receptor α. Am J Physiol Endocrinol Metab 2021; 320:E19-E29. [PMID: 33135461 DOI: 10.1152/ajpendo.00211.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Estetrol (E4), a natural estrogen synthesized by the human fetal liver, is currently evaluated in phase III clinical studies as a new menopause hormone therapy. Indeed, E4 significantly improves vasomotor and genito-urinary menopausal symptoms and prevents bone demineralization. Compared with other estrogens, E4 was found to have limited effects on coagulation factors in the liver of women allowing to expect less thrombotic events. To fully delineate its clinical potential, the aim of this study was to assess the effect of E4 on metabolic disorders. Here, we studied the pathophysiological consequences of a Western diet (42% kcal fat, 0.2% cholesterol) in ovariectomized female mice under chronic E4 treatment. We showed that E4 reduces body weight gain and improves glucose tolerance in both C57Bl/6 and LDLR-/- mice. To evaluate the role of hepatic estrogen receptor (ER) α in the preventive effect of E4 against obesity and associated disorders such as atherosclerosis and steatosis, mice harboring a hepatocyte-specific ERα deletion (LERKO) were crossed with LDLR-/- mice. Our results demonstrated that, whereas liver ERα is dispensable for the E4 beneficial actions on obesity and atheroma, it is necessary to prevent steatosis in mice. Overall, these findings suggest that E4 could prevent metabolic, hepatic, and vascular disorders occurring at menopause, extending the potential medical interest of this natural estrogen as a new hormonal treatment.NEW & NOTEWORTHY Estetrol prevents obesity, steatosis, and atherosclerosis in mice fed a Western diet. Hepatic ERα is necessary for the prevention of steatosis, but not of obesity and atherosclerosis.
Collapse
Affiliation(s)
- Mélissa Buscato
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), University of Toulouse, Toulouse, France
| | - Morgane Davezac
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), University of Toulouse, Toulouse, France
| | - Rana Zahreddine
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), University of Toulouse, Toulouse, France
| | - Marine Adlanmerini
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), University of Toulouse, Toulouse, France
| | - Raphaël Métivier
- CNRS, Univ Rennes, IGDR (Institut de Génétique De Rennes), Rennes, France
| | - Marianne Fillet
- Laboratory for the Analysis of Medicines, Center for Interdisciplinary Research on Medicines (CIRM), University of Liege, Quartier Hôpital, Liege, Belgium
| | - Gael Cobraiville
- Laboratory for the Analysis of Medicines, Center for Interdisciplinary Research on Medicines (CIRM), University of Liege, Quartier Hôpital, Liege, Belgium
| | - Cedric Moro
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), University of Toulouse, Toulouse, France
| | - Jean-Michel Foidart
- Groupe Interdisciplinaire de Génoprotéomique Appliquée, Université de Liège, Liège, Belgique
| | - Françoise Lenfant
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), University of Toulouse, Toulouse, France
| | - Pierre Gourdy
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), University of Toulouse, Toulouse, France
- Département de Diabétologie, Maladies Métaboliques et Nutrition, CHU de Toulouse, Toulouse, France
| | - Jean-François Arnal
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), University of Toulouse, Toulouse, France
| | - Coralie Fontaine
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), University of Toulouse, Toulouse, France
| |
Collapse
|
29
|
Mann SN, Hadad N, Nelson Holte M, Rothman AR, Sathiaseelan R, Ali Mondal S, Agbaga MP, Unnikrishnan A, Subramaniam M, Hawse J, Huffman DM, Freeman WM, Stout MB. Health benefits attributed to 17α-estradiol, a lifespan-extending compound, are mediated through estrogen receptor α. eLife 2020; 9:59616. [PMID: 33289482 PMCID: PMC7744101 DOI: 10.7554/elife.59616] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 12/07/2020] [Indexed: 02/06/2023] Open
Abstract
Metabolic dysfunction underlies several chronic diseases, many of which are exacerbated by obesity. Dietary interventions can reverse metabolic declines and slow aging, although compliance issues remain paramount. 17α-estradiol treatment improves metabolic parameters and slows aging in male mice. The mechanisms by which 17α-estradiol elicits these benefits remain unresolved. Herein, we show that 17α-estradiol elicits similar genomic binding and transcriptional activation through estrogen receptor α (ERα) to that of 17β-estradiol. In addition, we show that the ablation of ERα completely attenuates the beneficial metabolic effects of 17α-E2 in male mice. Our findings suggest that 17α-E2 may act through the liver and hypothalamus to improve metabolic parameters in male mice. Lastly, we also determined that 17α-E2 improves metabolic parameters in male rats, thereby proving that the beneficial effects of 17α-E2 are not limited to mice. Collectively, these studies suggest ERα may be a drug target for mitigating chronic diseases in male mammals.
Collapse
Affiliation(s)
- Shivani N Mann
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Oklahoma Center for Geroscience, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | - Niran Hadad
- The Jackson Laboratory, Bar Harbor, United States
| | - Molly Nelson Holte
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Alicia R Rothman
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | - Roshini Sathiaseelan
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | - Samim Ali Mondal
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | - Martin-Paul Agbaga
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | - Archana Unnikrishnan
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | | | - John Hawse
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Derek M Huffman
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, United States
| | - Willard M Freeman
- Oklahoma Center for Geroscience, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, United States.,Oklahoma City Veterans Affairs Medical Center, Oklahoma City, United States
| | - Michael B Stout
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Oklahoma Center for Geroscience, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| |
Collapse
|
30
|
DiStefano JK. NAFLD and NASH in Postmenopausal Women: Implications for Diagnosis and Treatment. Endocrinology 2020; 161:5890353. [PMID: 32776116 PMCID: PMC7473510 DOI: 10.1210/endocr/bqaa134] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 08/03/2020] [Indexed: 12/13/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) prevalence in women is increasing worldwide. Women of reproductive age have lower rates of NAFLD compared with men; however, this protection is lost following the menopausal transition when NAFLD prevalence in postmenopausal women becomes similar to or surpasses that in age-matched male counterparts. Ongoing epidemiological, clinical, and experimental studies indicate greater NAFLD risk and higher rates of severe hepatic fibrosis in postmenopausal women relative to premenopausal women, and that older women with NAFLD experience greater mortality than men. Investigations involving ovariectomized animal models demonstrate a causal relationship between estrogen deficiency and heightened susceptibility to the development of fatty liver and steatohepatitis, although dietary factors may exacerbate this complex relationship. The accumulated findings suggest that a better understanding of the interplay among menopausal status, metabolic comorbidities, and sex steroids in NAFLD pathogenesis is needed. Further, the mechanisms underlying the difference in NAFLD risk between postmenopausal and premenopausal women remain incompletely understood. The goals of this review are to summarize studies of NAFLD risk in postmenopausal women, discuss results from animal models of estrogen deficiency, and explore the development of NAFD within the context of altered sex hormone profiles resulting from the menopausal transition. Potential implications for the prevention, diagnosis, and treatment of NAFLD in this relatively understudied cohort are also addressed.
Collapse
Affiliation(s)
- Johanna K DiStefano
- Diabetes and Fibrotic Disease Research Unit, Translational Genomics Research Institute, Phoenix, Arizona
- Correspondence: Johanna K. DiStefano, Diabetes and Fibrotic Disease Research Unit, Translational Genomics Research Institute, Phoenix, AZ, USA. E-mail:
| |
Collapse
|
31
|
Della Torre S. Non-alcoholic Fatty Liver Disease as a Canonical Example of Metabolic Inflammatory-Based Liver Disease Showing a Sex-Specific Prevalence: Relevance of Estrogen Signaling. Front Endocrinol (Lausanne) 2020; 11:572490. [PMID: 33071979 PMCID: PMC7531579 DOI: 10.3389/fendo.2020.572490] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 08/20/2020] [Indexed: 12/11/2022] Open
Abstract
There is extensive evidence supporting the interplay between metabolism and immune response, that have evolved in close relationship, sharing regulatory molecules and signaling systems, to support biological functions. Nowadays, the disruption of this interaction in the context of obesity and overnutrition underlies the increasing incidence of many inflammatory-based metabolic diseases, even in a sex-specific fashion. During evolution, the interplay between metabolism and reproduction has reached a degree of complexity particularly high in female mammals, likely to ensure reproduction only under favorable conditions. Several factors may account for differences in the incidence and progression of inflammatory-based metabolic diseases between females and males, thus contributing to age-related disease development and difference in life expectancy between the two sexes. Among these factors, estrogens, acting mainly through Estrogen Receptors (ERs), have been reported to regulate several metabolic pathways and inflammatory processes particularly in the liver, the metabolic organ showing the highest degree of sexual dimorphism. This review aims to investigate on the interaction between metabolism and inflammation in the liver, focusing on the relevance of estrogen signaling in counteracting the development and progression of non-alcoholic fatty liver disease (NAFLD), a canonical example of metabolic inflammatory-based liver disease showing a sex-specific prevalence. Understanding the role of estrogens/ERs in the regulation of hepatic metabolism and inflammation may provide the basis for the development of sex-specific therapeutic strategies for the management of such an inflammatory-based metabolic disease and its cardio-metabolic consequences.
Collapse
Affiliation(s)
- Sara Della Torre
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| |
Collapse
|
32
|
De Courcy L, Bezak E, Marcu LG. Gender-dependent radiotherapy: The next step in personalised medicine? Crit Rev Oncol Hematol 2020; 147:102881. [PMID: 31991224 DOI: 10.1016/j.critrevonc.2020.102881] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 12/03/2019] [Accepted: 01/17/2020] [Indexed: 12/11/2022] Open
Abstract
Individuals do not react to radiation in a homogeneous manner. Recent radiogenomic research has proven that individual polymorphisms can correlate with treatment response most likely due to variation in the ability to recognise and repair DNA breaks. The difference in radiosensitivity between genders has been well documented, yet most radiotherapeutic guidelines are based solely on population averages rather than demographic subgroups such as age, race and gender. This paper is a review of the burgeoning literature available on the differences in efficacy and outcome of radiotherapy between genders. The work examines the effect of radiation on gender both from a tumour control as well as normal tissue toxicity perspective. While the literature reporting such findings is limited, the results show a small but significant difference in response to radiotherapy between sexes. Prospective and retrospective studies for evaluating these gender-specific differences are encouraged as a next step in personalised medicine.
Collapse
Affiliation(s)
- Louis De Courcy
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Eva Bezak
- Cancer Research Institute and School of Health Sciences, University of South Australia, Adelaide, SA, 5001, Australia; Department of Physics, University of Adelaide, North Terrace, Adelaide, SA, 5005, Australia
| | - Loredana G Marcu
- Cancer Research Institute and School of Health Sciences, University of South Australia, Adelaide, SA, 5001, Australia; Faculty of Informatics & Science, University of Oradea, Oradea, 410087, Romania.
| |
Collapse
|