1
|
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
|
2
|
Amar D, Gay NR, Jimenez-Morales D, Jean Beltran PM, Ramaker ME, Raja AN, Zhao B, Sun Y, Marwaha S, Gaul DA, Hershman SG, Ferrasse A, Xia A, Lanza I, Fernández FM, Montgomery SB, Hevener AL, Ashley EA, Walsh MJ, Sparks LM, Burant CF, Rector RS, Thyfault J, Wheeler MT, Goodpaster BH, Coen PM, Schenk S, Bodine SC, Lindholm ME. The mitochondrial multi-omic response to exercise training across rat tissues. Cell Metab 2024; 36:1411-1429.e10. [PMID: 38701776 PMCID: PMC11152996 DOI: 10.1016/j.cmet.2023.12.021] [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: 04/19/2023] [Revised: 09/27/2023] [Accepted: 12/15/2023] [Indexed: 05/05/2024]
Abstract
Mitochondria have diverse functions critical to whole-body metabolic homeostasis. Endurance training alters mitochondrial activity, but systematic characterization of these adaptations is lacking. Here, the Molecular Transducers of Physical Activity Consortium mapped the temporal, multi-omic changes in mitochondrial analytes across 19 tissues in male and female rats trained for 1, 2, 4, or 8 weeks. Training elicited substantial changes in the adrenal gland, brown adipose, colon, heart, and skeletal muscle. The colon showed non-linear response dynamics, whereas mitochondrial pathways were downregulated in brown adipose and adrenal tissues. Protein acetylation increased in the liver, with a shift in lipid metabolism, whereas oxidative proteins increased in striated muscles. Exercise-upregulated networks were downregulated in human diabetes and cirrhosis. Knockdown of the central network protein 17-beta-hydroxysteroid dehydrogenase 10 (HSD17B10) elevated oxygen consumption, indicative of metabolic stress. We provide a multi-omic, multi-tissue, temporal atlas of the mitochondrial response to exercise training and identify candidates linked to mitochondrial dysfunction.
Collapse
Affiliation(s)
- David Amar
- Stanford University, Stanford, CA, USA; Insitro, San Francisco, CA, USA
| | | | | | | | | | | | | | - Yifei Sun
- Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | | | - David A Gaul
- Georgia Institute of Technology, Atlanta, GA, USA
| | | | | | - Ashley Xia
- National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | | | - Martin J Walsh
- Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Lauren M Sparks
- Translational Research Institute AdventHealth, Orlando, FL, USA
| | | | | | - John Thyfault
- University of Kansas Medical Center, Kansas City, KS, USA
| | | | | | - Paul M Coen
- Translational Research Institute AdventHealth, Orlando, FL, USA
| | - Simon Schenk
- University of California, San Diego, La Jolla, CA, USA
| | - Sue C Bodine
- Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | | |
Collapse
|
3
|
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
|
4
|
Hernández-Saavedra D, Hinkley JM, Baer LA, Pinckard KM, Vidal P, Nirengi S, Brennan AM, Chen EY, Narain NR, Bussberg V, Tolstikov VV, Kiebish MA, Markunas C, Ilkayeva O, Goodpaster BH, Newgard CB, Goodyear LJ, Coen PM, Stanford KI. Chronic exercise improves hepatic acylcarnitine handling. iScience 2024; 27:109083. [PMID: 38361627 PMCID: PMC10867450 DOI: 10.1016/j.isci.2024.109083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 12/21/2023] [Accepted: 01/28/2024] [Indexed: 02/17/2024] Open
Abstract
Exercise mediates tissue metabolic function through direct and indirect adaptations to acylcarnitine (AC) metabolism, but the exact mechanisms are unclear. We found that circulating medium-chain acylcarnitines (AC) (C12-C16) are lower in active/endurance trained human subjects compared to sedentary controls, and this is correlated with elevated cardiorespiratory fitness and reduced adiposity. In mice, exercise reduced serum AC and increased liver AC, and this was accompanied by a marked increase in expression of genes involved in hepatic AC metabolism and mitochondrial β-oxidation. Primary hepatocytes from high-fat fed, exercise trained mice had increased basal respiration compared to hepatocytes from high-fat fed sedentary mice, which may be attributed to increased Ca2+ cycling and lipid uptake into mitochondria. The addition of specific medium- and long-chain AC to sedentary hepatocytes increased mitochondrial respiration, mirroring the exercise phenotype. These data indicate that AC redistribution is an exercise-induced mechanism to improve hepatic function and metabolism.
Collapse
Affiliation(s)
- Diego Hernández-Saavedra
- Dorothy M. Davis Heart and Lung Research Institute; Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - J. Matthew Hinkley
- AdventHealth Translational Research Institute, AdventHealth, Orlando, FL 32804, USA
| | - Lisa A. Baer
- Dorothy M. Davis Heart and Lung Research Institute; Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Kelsey M. Pinckard
- Dorothy M. Davis Heart and Lung Research Institute; Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Pablo Vidal
- Dorothy M. Davis Heart and Lung Research Institute; Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Shinsuke Nirengi
- Dorothy M. Davis Heart and Lung Research Institute; Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Andrea M. Brennan
- AdventHealth Translational Research Institute, AdventHealth, Orlando, FL 32804, USA
| | | | | | | | | | | | - Christina Markunas
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Departments of Pharmacology and Cancer Biology and Medicine, Durham, NC 27701, USA
| | - Olga Ilkayeva
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Departments of Pharmacology and Cancer Biology and Medicine, Durham, NC 27701, USA
| | - Bret H. Goodpaster
- AdventHealth Translational Research Institute, AdventHealth, Orlando, FL 32804, USA
| | - Christopher B. Newgard
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Departments of Pharmacology and Cancer Biology and Medicine, Durham, NC 27701, USA
| | - Laurie J. Goodyear
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Boston, MA 02215, USA
| | - Paul M. Coen
- AdventHealth Translational Research Institute, AdventHealth, Orlando, FL 32804, USA
| | - Kristin I. Stanford
- Dorothy M. Davis Heart and Lung Research Institute; Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
5
|
Gilley SP, Zarate MA, Zheng L, Jambal P, Yazza DN, Chintapalli SV, MacLean PS, Wright CJ, Rozance PJ, Shankar K. Metabolic and fecal microbial changes in adult fetal growth restricted mice. Pediatr Res 2024; 95:647-659. [PMID: 37935884 PMCID: PMC10899111 DOI: 10.1038/s41390-023-02869-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 09/13/2023] [Accepted: 10/12/2023] [Indexed: 11/09/2023]
Abstract
BACKGROUND Fetal growth restriction (FGR) increases risk for development of obesity and type 2 diabetes. Using a mouse model of FGR, we tested whether metabolic outcomes were exacerbated by high-fat diet challenge or associated with fecal microbial taxa. METHODS FGR was induced by maternal calorie restriction from gestation day 9 to 19. Control and FGR offspring were weaned to control (CON) or 45% fat diet (HFD). At age 16 weeks, offspring underwent intraperitoneal glucose tolerance testing, quantitative MRI body composition assessment, and energy balance studies. Total microbial DNA was used for amplification of the V4 variable region of the 16 S rRNA gene. Multivariable associations between groups and genera abundance were assessed using MaAsLin2. RESULTS Adult male FGR mice fed HFD gained weight faster and had impaired glucose tolerance compared to control HFD males, without differences among females. Irrespective of weaning diet, adult FGR males had depletion of Akkermansia, a mucin-residing genus known to be associated with weight gain and glucose handling. FGR females had diminished Bifidobacterium. Metabolic changes in FGR offspring were associated with persistent gut microbial changes. CONCLUSION FGR results in persistent gut microbial dysbiosis that may be a therapeutic target to improve metabolic outcomes. IMPACT Fetal growth restriction increases risk for metabolic syndrome later in life, especially if followed by rapid postnatal weight gain. We report that a high fat diet impacts weight and glucose handling in a mouse model of fetal growth restriction in a sexually dimorphic manner. Adult growth-restricted offspring had persistent changes in fecal microbial taxa known to be associated with weight, glucose homeostasis, and bile acid metabolism, particularly Akkermansia, Bilophilia and Bifidobacteria. The gut microbiome may represent a therapeutic target to improve long-term metabolic outcomes related to fetal growth restriction.
Collapse
Affiliation(s)
- Stephanie P Gilley
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, CO, USA.
| | - Miguel A Zarate
- Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Lijun Zheng
- Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Purevsuren Jambal
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, CO, USA
| | - Deaunabah N Yazza
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, CO, USA
| | - Sree V Chintapalli
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Paul S MacLean
- Department of Medicine, Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Clyde J Wright
- Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Paul J Rozance
- Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Kartik Shankar
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
6
|
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
|
7
|
Tian Y, Hong X, Xie Y, Guo Z, Yu Q. 17β-Estradiol (E 2) Upregulates the ERα/SIRT1/PGC-1α Signaling Pathway and Protects Mitochondrial Function to Prevent Bilateral Oophorectomy (OVX)-Induced Nonalcoholic Fatty Liver Disease (NAFLD). Antioxidants (Basel) 2023; 12:2100. [PMID: 38136219 PMCID: PMC10740447 DOI: 10.3390/antiox12122100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/30/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
Premature menopause is associated with an increased prevalence of nonalcoholic fatty liver disease (NAFLD). Menopausal hormone therapy (MHT) has been widely used in clinical practice and has the potential to protect mitochondrial function and alleviate NAFLD. After bilateral oophorectomy (OVX), female rats without 17β-estradiol (E2) intervention developed NAFLD, whereas E2 supplementation was effective in preventing NAFLD in female rats. The altered pathways and cellular events from both comparison pairs, namely, the OVX vs. sham group and the OVX vs. E2 group, were assessed using transcriptomic analysis. KEGG pathways enriched by both transcriptomic and metabolomic analyses strongly suggest that oxidative phosphorylation is a vital pathway that changes during the development of NAFLD and remains unchanged when E2 is applied. Liver tissue from the OVX-induced NAFLD group exhibited increased lipid peroxidation, impaired mitochondria, and downregulated ERα/SIRT1/PGC-1α expression. An in vitro study indicated that the protective effect of E2 treatment on hepatic steatosis could be abolished when ERα or SIRT1 was selectively inhibited. This damage was accompanied by reduced mitochondrial complex activity and increased lipid peroxidation. The current research indicates that E2 upregulates the ERα/SIRT1/PGC-1α signaling pathway and protects mitochondrial function to prevent OVX-induced NAFLD.
Collapse
Affiliation(s)
| | | | | | | | - Qi Yu
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetric & Gynecologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Peking Union Medical College Hospital (Dongdan Campus), No.1 Shuaifuyuan Wangfujing Dongcheng District, Beijing 100730, China; (Y.T.); (X.H.); (Y.X.); (Z.G.)
| |
Collapse
|
8
|
Kugler BA, Cao X, Wenger M, Franczak E, McCoin CS, Von Schulze A, Morris EM, Thyfault JP. Divergence in aerobic capacity influences hepatic and systemic metabolic adaptations to bile acid sequestrant and short-term high-fat/sucrose feeding in rats. Am J Physiol Regul Integr Comp Physiol 2023; 325:R712-R724. [PMID: 37811712 PMCID: PMC11178297 DOI: 10.1152/ajpregu.00133.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 09/29/2023] [Accepted: 10/03/2023] [Indexed: 10/10/2023]
Abstract
High versus low aerobic capacity significantly impacts the risk for metabolic diseases. Rats selectively bred for high or low intrinsic aerobic capacity differently modify hepatic bile acid metabolism in response to high-fat diets (HFDs). Here we tested if a bile acid sequestrant would alter hepatic and whole body metabolism differently in rats with high and low aerobic capacity fed a 1-wk HFD. Male rats (8 mo of age) that were artificially selected to be high (HCR) and low-capacity runners (LCR) with divergent intrinsic aerobic capacities were transitioned from a low-fat diet (LFD, 10% fat) to an HFD (45% fat) with or without a bile acid sequestrant (BA-Seq, 2% cholestyramine resin) for 7 days while maintained in an indirect calorimetry system. HFD + BA-Seq increased fecal excretion of lipids and bile acids and prevented weight and fat mass gain in both strains. Interestingly, HCR rats had increased adaptability to enhance fecal bile acid and lipid loss, resulting in more significant energy loss than their LCR counterpart. In addition, BA-Seq induced a greater expression of hepatic CYP7A1 gene expression, the rate-limiting enzyme of bile acid synthesis in HCR rats both on HFD and HFD + BA-Seq diets. HCR displayed a more significant reduction of RQ in response to HFD than LCR, but HFD + BA-Seq lowered RQ in both groups compared with HFD alone, demonstrating a pronounced impact on metabolic flexibility. In conclusion, BA-Seq provides uniform metabolic benefits for metabolic flexibility and adiposity, but rats with higher aerobic capacity display adaptability for hepatic bile acid metabolism.NEW & NOTEWORTHY The administration of bile acid sequestrant (BA-Seq) has uniform metabolic benefits in terms of metabolic flexibility and adiposity in rats with high and low aerobic capacity. However, rats with higher aerobic capacity demonstrate greater adaptability in hepatic bile acid metabolism, resulting in increased fecal bile acid and lipid loss, as well as enhanced fecal energy loss.
Collapse
Affiliation(s)
- Benjamin A Kugler
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas Center for Metabolism and Obesity Research, Kansas City, Missouri, United States
- Department of Internal Medicine, Division of Endocrinology and Metabolism, KU Diabetes Institute, The University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Xin Cao
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas Center for Metabolism and Obesity Research, Kansas City, Missouri, United States
- Department of Internal Medicine, Division of Endocrinology and Metabolism, KU Diabetes Institute, The University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Madi Wenger
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas Center for Metabolism and Obesity Research, Kansas City, Missouri, United States
- Department of Internal Medicine, Division of Endocrinology and Metabolism, KU Diabetes Institute, The University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Edziu Franczak
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas Center for Metabolism and Obesity Research, Kansas City, Missouri, United States
- Department of Internal Medicine, Division of Endocrinology and Metabolism, KU Diabetes Institute, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas City Veterans Affairs Medical Center, Kansas City, Missouri, United States
| | - Colin S McCoin
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas Center for Metabolism and Obesity Research, Kansas City, Missouri, United States
- Center for Children's Healthy Lifestyles and Nutrition, Kansas City, Missouri, United States
- Department of Internal Medicine, Division of Endocrinology and Metabolism, KU Diabetes Institute, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas City Veterans Affairs Medical Center, Kansas City, Missouri, United States
| | - Alex Von Schulze
- Stowers Research Institute, Kansas City, Missouri, United States
| | - E Matthew Morris
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas Center for Metabolism and Obesity Research, Kansas City, Missouri, United States
- Center for Children's Healthy Lifestyles and Nutrition, Kansas City, Missouri, United States
- Department of Internal Medicine, Division of Endocrinology and Metabolism, KU Diabetes Institute, The University of Kansas Medical Center, Kansas City, Kansas, United States
| | - John P Thyfault
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas Center for Metabolism and Obesity Research, Kansas City, Missouri, United States
- Center for Children's Healthy Lifestyles and Nutrition, Kansas City, Missouri, United States
- Department of Internal Medicine, Division of Endocrinology and Metabolism, KU Diabetes Institute, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas City Veterans Affairs Medical Center, Kansas City, Missouri, United States
| |
Collapse
|
9
|
Ruebel ML, Borengasser SJ, Zhong Y, Kang P, Faske J, Shankar K. Maternal Exercise Prior to and during Gestation Induces Sex-Specific Alterations in the Mouse Placenta. Int J Mol Sci 2023; 24:16441. [PMID: 38003633 PMCID: PMC10671464 DOI: 10.3390/ijms242216441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/08/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
While exercise (EX) during pregnancy is beneficial for both mother and child, little is known about the mechanisms by which maternal exercise mediates changes in utero. Six-week-old female C57BL/6 mice were divided into two groups: with (exercise, EX; N = 7) or without (sedentary, SED; N = 8) access to voluntary running wheels. EX was provided via 24 h access to wheels for 10 weeks prior to conception until late pregnancy (18.5 days post coitum). Sex-stratified placentas and fetal livers were collected. Microarray analysis of SED and EX placentas revealed that EX affected gene transcript expression of 283 and 661 transcripts in male and female placentas, respectively (±1.4-fold, p < 0.05). Gene Set Enrichment and Ingenuity Pathway Analyses of male placentas showed that EX led to inhibition of signaling pathways, biological functions, and down-regulation of transcripts related to lipid and steroid metabolism, while EX in female placentas led to activation of pathways, biological functions, and gene expression related to muscle growth, brain, vascular development, and growth factors. Overall, our results suggest that the effects of maternal EX on the placenta and presumably on the offspring are sexually dimorphic.
Collapse
Affiliation(s)
- Meghan L. Ruebel
- Microbiome and Metabolism Research Unit, USDA-ARS, Southeast Area, Little Rock, AR 72202, USA;
- Arkansas Children’s Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA; (Y.Z.); (J.F.)
| | - Sarah J. Borengasser
- Tobacco Settlement Endowment Trust Health Promotion Research Center, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Pediatrics—Endocrinology & Diabetes, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Ying Zhong
- Arkansas Children’s Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA; (Y.Z.); (J.F.)
| | - Ping Kang
- Arkansas Children’s Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA; (Y.Z.); (J.F.)
| | - Jennifer Faske
- Arkansas Children’s Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA; (Y.Z.); (J.F.)
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA
| | - Kartik Shankar
- Department of Pediatrics, Section of Nutrition, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
10
|
Wang K, Moore A, Grayson C, Mailloux RJ. S-nitroso-glutathione (GSNO) inhibits hydrogen peroxide production by alpha-ketoglutarate dehydrogenase: An investigation into sex and diet effects. Free Radic Biol Med 2023; 204:287-300. [PMID: 37225107 DOI: 10.1016/j.freeradbiomed.2023.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/06/2023] [Accepted: 05/11/2023] [Indexed: 05/26/2023]
Abstract
Pyruvate dehydrogenase (PDH) and α-ketoglutarate dehydrogenase (KGDH) are vital sources of hydrogen peroxide (H2O2) and key sites for redox regulation. Here, we report KGDH is more sensitive to inhibition by S-nitroso-glutathione (GSNO) when compared to PDH and deactivation of both enzymes by nitro modification is affected by sex and diet. Liver mitochondria from male C57BL/6N mice displayed a robust inhibition of H2O2 production after exposure to 500-2000 μM GSNO. H2O2 genesis by PDH was not significantly affected by GSNO. Purified KGDH of porcine heart origin displayed a ∼82% decrease in H2O2 generating activity at 500 μM GSNO, which was mirrored by a decrease in NADH production. By contrast, H2O2- and NADH-producing activity of purified PDH was only minimally affected by an incubation in 500 μM GSNO. Incubations in GSNO had no significant effect on the H2O2-generating activity of KGDH and PDH in female liver mitochondria when compared to samples collected from males, which was attributed to higher GSNO reductase (GSNOR) activity. High fat feeding augmented the GSNO-mediated inhibition of KGDH in liver mitochondria from male mice. Exposure of male mice to a HFD also resulted in a significant decrease in the GSNO-mediated inhibition of H2O2 genesis by PDH, an effect not observed in mice fed a control-matched diet (CD). Female mice displayed higher resistance to the GSNO-induced inhibition of H2O2 production, regardless of being fed a CD or HFD. However, exposure to a HFD did result in a small but significant decrease in H2O2 production by KGDH and PDH when female liver mitochondria were treated with GSNO. Although, the effect was less when compared to their male counterparts. Collectively, we show for the first time GSNO deactivates H2O2 production by α-keto acid dehydrogenases and we demonstrate that sex and diet are determinants for the nitro-inhibition of both KGDH and PDH.
Collapse
Affiliation(s)
- Kevin Wang
- The School of Human Nutrition, Faculty of Agricultural and Environmental Sciences, McGill University, Ste.-Anne-de-Bellevue, Quebec, Canada
| | - Amanda Moore
- The School of Human Nutrition, Faculty of Agricultural and Environmental Sciences, McGill University, Ste.-Anne-de-Bellevue, Quebec, Canada
| | - Cathryn Grayson
- The School of Human Nutrition, Faculty of Agricultural and Environmental Sciences, McGill University, Ste.-Anne-de-Bellevue, Quebec, Canada
| | - Ryan J Mailloux
- The School of Human Nutrition, Faculty of Agricultural and Environmental Sciences, McGill University, Ste.-Anne-de-Bellevue, Quebec, Canada.
| |
Collapse
|
11
|
Undamatla R, Fagunloye OG, Chen J, Edmunds LR, Murali A, Mills A, Xie B, Pangburn MM, Sipula I, Gibson G, St Croix C, Jurczak MJ. Reduced mitophagy is an early feature of NAFLD and liver-specific PARKIN knockout hastens the onset of steatosis, inflammation and fibrosis. Sci Rep 2023; 13:7575. [PMID: 37165006 PMCID: PMC10172344 DOI: 10.1038/s41598-023-34710-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 05/05/2023] [Indexed: 05/12/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) encompasses a spectrum of pathologies that includes steatosis, steatohepatitis (NASH) and fibrosis and is strongly associated with insulin resistance and type 2 diabetes. Changes in mitochondrial function are implicated in the pathogenesis of NAFLD, particularly in the transition from steatosis to NASH. Mitophagy is a mitochondrial quality control mechanism that allows for the selective removal of damaged mitochondria from the cell via the autophagy pathway. While past work demonstrated a negative association between liver fat content and rates of mitophagy, when changes in mitophagy occur during the pathogenesis of NAFLD and whether such changes contribute to the primary endpoints associated with the disease are currently poorly defined. We therefore undertook the studies described here to establish when alterations in mitophagy occur during the pathogenesis of NAFLD, as well as to determine the effects of genetic inhibition of mitophagy via conditional deletion of a key mitophagy regulator, PARKIN, on the development of steatosis, insulin resistance, inflammation and fibrosis. We find that loss of mitophagy occurs early in the pathogenesis of NAFLD and that loss of PARKIN accelerates the onset of key NAFLD disease features. These observations suggest that loss of mitochondrial quality control in response to nutritional stress may contribute to mitochondrial dysfunction and the pathogenesis of NAFLD.
Collapse
Affiliation(s)
- R Undamatla
- Division of Endocrinology and Metabolism, Department of Medicine, School of Medicine, University of Pittsburgh, 200 Lothrop Street, BST W1060, Pittsburgh, PA, 15213, USA
| | - O G Fagunloye
- Division of Endocrinology and Metabolism, Department of Medicine, School of Medicine, University of Pittsburgh, 200 Lothrop Street, BST W1060, Pittsburgh, PA, 15213, USA
| | - J Chen
- Division of Endocrinology and Metabolism, Department of Medicine, School of Medicine, University of Pittsburgh, 200 Lothrop Street, BST W1060, Pittsburgh, PA, 15213, USA
| | - L R Edmunds
- Division of Endocrinology and Metabolism, Department of Medicine, School of Medicine, University of Pittsburgh, 200 Lothrop Street, BST W1060, Pittsburgh, PA, 15213, USA
| | - A Murali
- Division of Endocrinology and Metabolism, Department of Medicine, School of Medicine, University of Pittsburgh, 200 Lothrop Street, BST W1060, Pittsburgh, PA, 15213, USA
| | - A Mills
- Division of Endocrinology and Metabolism, Department of Medicine, School of Medicine, University of Pittsburgh, 200 Lothrop Street, BST W1060, Pittsburgh, PA, 15213, USA
| | - B Xie
- Division of Endocrinology and Metabolism, Department of Medicine, School of Medicine, University of Pittsburgh, 200 Lothrop Street, BST W1060, Pittsburgh, PA, 15213, USA
| | - M M Pangburn
- Division of Endocrinology and Metabolism, Department of Medicine, School of Medicine, University of Pittsburgh, 200 Lothrop Street, BST W1060, Pittsburgh, PA, 15213, USA
| | - I Sipula
- Division of Endocrinology and Metabolism, Department of Medicine, School of Medicine, University of Pittsburgh, 200 Lothrop Street, BST W1060, Pittsburgh, PA, 15213, USA
| | - G Gibson
- Department of Cell Biology, Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, PA, USA
| | - C St Croix
- Department of Cell Biology, Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, PA, USA
| | - M J Jurczak
- Division of Endocrinology and Metabolism, Department of Medicine, School of Medicine, University of Pittsburgh, 200 Lothrop Street, BST W1060, Pittsburgh, PA, 15213, USA.
- Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
12
|
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
|
13
|
Witte S, Boshnakovska A, Özdemir M, Chowdhury A, Rehling P, Aich A. Defective COX1 expression in aging mice liver. Biol Open 2023; 12:292575. [PMID: 36861685 PMCID: PMC10003073 DOI: 10.1242/bio.059844] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 01/26/2023] [Indexed: 03/03/2023] Open
Abstract
Mitochondrial defects are associated with aging processes and age-related diseases, including cardiovascular diseases, neurodegenerative diseases and cancer. In addition, some recent studies suggest mild mitochondrial dysfunctions appear to be associated with longer lifespans. In this context, liver tissue is considered to be largely resilient to aging and mitochondrial dysfunction. Yet, in recent years studies report dysregulation of mitochondrial function and nutrient sensing pathways in ageing livers. Therefore, we analyzed the effects of the aging process on mitochondrial gene expression in liver using wildtype C57BL/6N mice. In our analyses, we observed alteration in mitochondrial energy metabolism with age. To assess if defects in mitochondrial gene expression are linked to this decline, we applied a Nanopore sequencing based approach for mitochondrial transcriptomics. Our analyses show that a decrease of the Cox1 transcript correlates with reduced respiratory complex IV activity in older mice livers.
Collapse
Affiliation(s)
- Steffen Witte
- Department of Cellular Biochemistry, University Medical Center, Göttingen, 37073, Germany
| | - Angela Boshnakovska
- Department of Cellular Biochemistry, University Medical Center, Göttingen, 37073, Germany
| | - Metin Özdemir
- Department of Cellular Biochemistry, University Medical Center, Göttingen, 37073, Germany
| | - Arpita Chowdhury
- Department of Cellular Biochemistry, University Medical Center, Göttingen, 37073, Germany
| | - Peter Rehling
- Department of Cellular Biochemistry, University Medical Center, Göttingen, 37073, Germany.,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, 37075, Germany.,Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Translational Neuroinflammation and Automated Microscopy, Göttingen, 37075, Germany.,Max Planck Institute for Multidisciplinary Sciences, Göttingen, 37077, Germany
| | - Abhishek Aich
- Department of Cellular Biochemistry, University Medical Center, Göttingen, 37073, Germany.,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, 37075, Germany
| |
Collapse
|
14
|
Kugler BA, Thyfault JP, McCoin CS. Sexually dimorphic hepatic mitochondrial adaptations to exercise: a mini-review. J Appl Physiol (1985) 2023; 134:685-691. [PMID: 36701482 PMCID: PMC10027083 DOI: 10.1152/japplphysiol.00711.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/23/2023] [Accepted: 01/23/2023] [Indexed: 01/27/2023] Open
Abstract
Exercise is a physiological stress that disrupts tissue and cellular homeostasis while enhancing systemic metabolic energy demand mainly through the increased workload of skeletal muscle. Although the extensive focus has been on skeletal muscle adaptations to exercise, the liver senses these disruptions in metabolic energy homeostasis and responds to provide the required substrates to sustain increased demand. Hepatic metabolic flexibility is an energetically costly process that requires continuous mitochondrial production of the cellular currency ATP. To do so, the liver must maintain a healthy functioning mitochondrial pool, attained through well-regulated and dynamic processes. Intriguingly, some of these responses are sex-dependent. This mini-review examines the hepatic mitochondrial adaptations to exercise with a focus on sexual dimorphism.
Collapse
Affiliation(s)
- Benjamin A Kugler
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, Kansas City, Kansas, United States
- KU Diabetes Institute, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas Center for Metabolism and Obesity Research, The University of Kansas Medical Center, Kansas City, Kansas, United States
| | - John P Thyfault
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, Kansas City, Kansas, United States
- KU Diabetes Institute, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas Center for Metabolism and Obesity Research, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Center for Children's Healthy Lifestyles and Nutrition, Kansas City, Missouri, United States
- Division of Endocrinology and Metabolism, Department of Internal Medicine, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas City Veterans Affairs Medical Center, Kansas City, Missouri, United States
| | - Colin S McCoin
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, Kansas City, Kansas, United States
- KU Diabetes Institute, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas Center for Metabolism and Obesity Research, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Center for Children's Healthy Lifestyles and Nutrition, Kansas City, Missouri, United States
- Kansas City Veterans Affairs Medical Center, Kansas City, Missouri, United States
| |
Collapse
|
15
|
Undamatla R, Fagunloye OG, Chen J, Edmunds LR, Murali A, Mills A, Xie B, Pangburn MM, Sipula I, Gibson G, Croix CS, Jurczak MJ. Reduced hepatocyte mitophagy is an early feature of NAFLD pathogenesis and hastens the onset of steatosis, inflammation and fibrosis. RESEARCH SQUARE 2023:rs.3.rs-2469234. [PMID: 36711642 PMCID: PMC9882688 DOI: 10.21203/rs.3.rs-2469234/v1] [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: 01/18/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) encompasses a spectrum of pathologies that includes steatosis, steatohepatitis (NASH) and fibrosis and is strongly associated with insulin resistance and type 2 diabetes. Changes in mitochondrial function are implicated in the pathogenesis of NAFLD, particularly in the transition from steatosis to NASH. Mitophagy is a mitochondrial quality control mechanism that allows for the selective removal of damaged mitochondria from the cell via the autophagy pathway. While past work demonstrated a negative association between liver fat content and rates of mitophagy, when changes in mitophagy occur during the pathogenesis of NAFLD and whether such changes contribute to the primary endpoints associated with the disease are currently poorly defined. We therefore undertook the studies described here to establish when alterations in mitophagy occur during the pathogenesis of NAFLD, as well as to determine the effects of genetic inhibition of mitophagy via conditional deletion of a key mitophagy regulator, PARKIN, on the development of steatosis, insulin resistance, inflammation and fibrosis. We find that loss of mitophagy occurs early in the pathogenesis of NAFLD and that loss of PARKIN hastens the onset but not severity of key NAFLD disease features. These observations suggest that loss of mitochondrial quality control in response to nutritional stress may contribute to mitochondrial dysfunction and the pathogenesis of NAFLD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Ian Sipula
- University of Pittsburgh School of Medicine
| | | | | | | |
Collapse
|
16
|
Amar D, Gay NR, Jimenez-Morales D, Beltran PMJ, Ramaker ME, Raja AN, Zhao B, Sun Y, Marwaha S, Gaul D, Hershman SG, Xia A, Lanza I, Fernandez FM, Montgomery SB, Hevener AL, Ashley EA, Walsh MJ, Sparks LM, Burant CF, Rector RS, Thyfault J, Wheeler MT, Goodpaster BH, Coen PM, Schenk S, Bodine SC, Lindholm ME. The mitochondrial multi-omic response to exercise training across tissues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.13.523698. [PMID: 36711881 PMCID: PMC9882193 DOI: 10.1101/2023.01.13.523698] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Mitochondria are adaptable organelles with diverse cellular functions critical to whole-body metabolic homeostasis. While chronic endurance exercise training is known to alter mitochondrial activity, these adaptations have not yet been systematically characterized. Here, the Molecular Transducers of Physical Activity Consortium (MoTrPAC) mapped the longitudinal, multi-omic changes in mitochondrial analytes across 19 tissues in male and female rats endurance trained for 1, 2, 4 or 8 weeks. Training elicited substantial changes in the adrenal gland, brown adipose, colon, heart and skeletal muscle, while we detected mild responses in the brain, lung, small intestine and testes. The colon response was characterized by non-linear dynamics that resulted in upregulation of mitochondrial function that was more prominent in females. Brown adipose and adrenal tissues were characterized by substantial downregulation of mitochondrial pathways. Training induced a previously unrecognized robust upregulation of mitochondrial protein abundance and acetylation in the liver, and a concomitant shift in lipid metabolism. The striated muscles demonstrated a highly coordinated response to increase oxidative capacity, with the majority of changes occurring in protein abundance and post-translational modifications. We identified exercise upregulated networks that are downregulated in human type 2 diabetes and liver cirrhosis. In both cases HSD17B10, a central dehydrogenase in multiple metabolic pathways and mitochondrial tRNA maturation, was the main hub. In summary, we provide a multi-omic, cross-tissue atlas of the mitochondrial response to training and identify candidates for prevention of disease-associated mitochondrial dysfunction.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yifei Sun
- Icahn School of Medicine at Mount Sinai, New York City, NY
| | | | | | | | - Ashley Xia
- National Institutes of Health, Bethesda, MD
| | | | | | | | | | | | - Martin J Walsh
- Icahn School of Medicine at Mount Sinai, New York City, NY
| | - Lauren M Sparks
- AdventHealth Translational Research Institute for Metabolism and Diabetes, Orlando, FL
| | | | | | - John Thyfault
- University of Kansas Medical Center, Kansas City, KS
| | | | - Bret H. Goodpaster
- AdventHealth Translational Research Institute for Metabolism and Diabetes, Orlando, FL
| | - Paul M. Coen
- AdventHealth Translational Research Institute for Metabolism and Diabetes, Orlando, FL
| | - Simon Schenk
- University of California, San Diego, La Jolla, CA
| | | | | | | |
Collapse
|
17
|
Sex Differences of Radiation Damage in High-Fat-Diet-Fed Mice and the Regulatory Effect of Melatonin. Nutrients 2022; 15:nu15010064. [PMID: 36615722 PMCID: PMC9823527 DOI: 10.3390/nu15010064] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/18/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022] Open
Abstract
The consumption of a high-fat diet (HFD) and exposure to ionizing radiation (IR) are closely associated with many diseases. To evaluate the interaction between HFDs and IR-induced injury, we gave mice whole abdominal irradiation (WAI) to examine the extent of intestinal injury under different dietary conditions. Melatonin (MLT) is a free radical scavenger that effectively prevents hematopoietic, immune, and gastrointestinal damage induced by IR. However, its effects on WAI-induced intestinal injury in HFD-fed mice remain unclear. We demonstrated that MLT can promote intestinal structural repair following WAI and enhance the regeneration capacity of Lgr5+ intestinal stem cells. In addition, we investigated the effects of radiation damage on sexual dimorphism in HFD-fed mice. The results showed that the degree of IR-induced intestinal injury was more severe in the HFD-fed female mice. MLT preserved the intestinal microbiota composition of HFD-fed mice and increased the abundance of Bacteroides and Proteobacteria in male and female mice, respectively. In conclusion, MLT may reduce the negative effects of HFD and IR, thereby providing assistance in preserving the structure and function of the intestine.
Collapse
|
18
|
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
|
19
|
Hughey CC, Puchalska P, Crawford PA. Integrating the contributions of mitochondrial oxidative metabolism to lipotoxicity and inflammation in NAFLD pathogenesis. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159209. [DOI: 10.1016/j.bbalip.2022.159209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 06/25/2022] [Accepted: 07/27/2022] [Indexed: 11/28/2022]
|
20
|
Stierwalt HD, Morris EM, Maurer A, Apte U, Phillips K, Li T, Meers GME, Koch LG, Britton SL, Graf G, Rector RS, Mercer K, Shankar K, Thyfault JP. Rats with high aerobic capacity display enhanced transcriptional adaptability and upregulation of bile acid metabolism in response to an acute high-fat diet. Physiol Rep 2022; 10:e15405. [PMID: 35923133 PMCID: PMC9350427 DOI: 10.14814/phy2.15405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 07/01/2022] [Accepted: 07/13/2022] [Indexed: 06/09/2023] Open
Abstract
Rats selectively bred for the high intrinsic aerobic capacity runner (HCR) or low aerobic capacity runner (LCR) show pronounced differences in susceptibility for high-fat/high sucrose (HFHS) diet-induced hepatic steatosis and insulin resistance, replicating the protective effect of high aerobic capacity in humans. We have previously shown multiple systemic differences in energy and substrate metabolism that impacts steatosis between HCR and LCR rats. This study aimed to investigate hepatic-specific mechanisms of action via changes in gene transcription. Livers of HCR rats had a greater number of genes that significantly changed in response to 3-day HFHS compared with LCR rats (171 vs. 75 genes: >1.5-fold, p < 0.05). HCR and LCR rats displayed numerous baseline differences in gene expression while on a low-fat control diet (CON). A 3-day HFHS diet resulted in greater expression of genes involved in the conversion of excess acetyl-CoA to cholesterol and bile acid (BA) synthesis compared with the CON diet in HCR, but not LCR rats. These results were associated with higher fecal BA loss and lower serum BA concentrations in HCR rats. Exercise studies in rats and mice also revealed higher hepatic expression of cholesterol and BA synthesis genes. Overall, these results suggest that high aerobic capacity and exercise are associated with upregulated BA synthesis paired with greater fecal excretion of cholesterol and BA, an effect that may play a role in protection against hepatic steatosis in rodents.
Collapse
Affiliation(s)
- Harrison D. Stierwalt
- Molecular and Integrative PhysiologyUniversity of Kansas Medical CenterKansas CityMissouriUSA
- Research ServiceKansas City VA Medical CenterKansas CityMissouriUSA
| | - E. Matthew Morris
- Molecular and Integrative PhysiologyUniversity of Kansas Medical CenterKansas CityMissouriUSA
| | - Adrianna Maurer
- Molecular and Integrative PhysiologyUniversity of Kansas Medical CenterKansas CityMissouriUSA
| | - Udayan Apte
- Department of Pharmacology, Toxicology, and TherapeuticsUniversity of Kansas Medical CenterKansas CityMissouriUSA
| | | | - Tiangang Li
- Department of PhysiologyUniversity of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
| | - Grace M. E. Meers
- Division of Gastroenterology and HepatologyUniversity of MissouriColumbiaMissouriUSA
- Division of Nutrition and Exercise PhysiologyColumbiaMissouriUSA
| | - Lauren G. Koch
- Physiology and PharmacologyThe University of ToledoToledoOhioUSA
| | | | - Greg Graf
- Department of Pharmaceutical SciencesSaha Cardiovascular Research Center, University of KentuckyLexingtonKentuckyUSA
| | - R. Scott Rector
- Division of Gastroenterology and HepatologyUniversity of MissouriColumbiaMissouriUSA
- Division of Nutrition and Exercise PhysiologyColumbiaMissouriUSA
- Research ServiceHarry S Truman Memorial VA HospitalColumbiaMissouriUSA
| | - Kelly Mercer
- Arkansas Children's Nutrition CenterUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Kartik Shankar
- Section of Nutrition, Department of PediatricsUniversity of Colorado School of Medicine Anschutz Medical CampusAuroraColoradoUSA
| | - John P. Thyfault
- Molecular and Integrative PhysiologyUniversity of Kansas Medical CenterKansas CityMissouriUSA
- Research ServiceKansas City VA Medical CenterKansas CityMissouriUSA
| |
Collapse
|
21
|
Cunningham RP, Moore MP, Dashek RJ, Meers GM, Jepkemoi V, Takahashi T, Vieira-Potter VJ, Kanaley JA, Booth FW, Rector RS. Hepatocyte-specific eNOS deletion impairs exercise-induced adaptations in hepatic mitochondrial function and autophagy. Obesity (Silver Spring) 2022; 30:1066-1078. [PMID: 35357089 PMCID: PMC9050943 DOI: 10.1002/oby.23414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/24/2022] [Accepted: 02/11/2022] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Endothelial nitric oxide synthase (eNOS) is a potential mediator of exercise-induced hepatic mitochondrial adaptations. METHODS Here, male and female hepatocyte-specific eNOS knockout (eNOShep-/- ) and intact hepatic eNOS (eNOSfl/fl ) mice performed voluntary wheel-running exercise (EX) or remained in sedentary cage conditions for 10 weeks. RESULTS EX resolved the exacerbated hepatic steatosis in eNOShep-/- male mice. Elevated hydrogen peroxide emission (~50% higher in eNOShep-/- vs. eNOSfl/fl mice) was completely ablated with EX. Interestingly, EX increased [1-14 C] palmitate oxidation in eNOSfl/fl male mice, but this was blunted in the eNOShep-/- male mice. eNOShep-/- mice had lower markers of the energy sensors AMP-activated protein kinase (AMPK)/phospho- (p)AMPK and mammalian target of rapamycin (mTOR) and p-mTOR, as well as the autophagy initiators serine/threonine-protein kinase ULK1 and pULK1, compared with eNOSfl/fl mice. Females showed elevated electron transport chain protein content and markers of mitochondrial biogenesis (transcription factor A, mitochondrial, peroxisome proliferator-activated receptor-gamma coactivator 1α). CONCLUSIONS Collectively, this study demonstrates for the first time, to the authors' knowledge, the requirement of eNOS in hepatocytes in the EX-induced increases in hepatic fatty acid oxidation in male mice. Deletion of eNOS in hepatocytes also appears to impair the energy-sensing ability of the cell and inhibit the activation of the autophagy initiating factor ULK1. These data uncover the important and novel role of hepatocyte eNOS in EX-induced hepatic mitochondrial adaptations.
Collapse
Affiliation(s)
- Rory P. Cunningham
- Research Service, Harry S Truman Memorial Veterans Medical Center, Columbia, Missouri 65212, USA
- Nutrition and Exercise Physiology, University of Missouri, Columbia, MO 65211, USA
| | - Mary P. Moore
- Research Service, Harry S Truman Memorial Veterans Medical Center, Columbia, Missouri 65212, USA
- Nutrition and Exercise Physiology, University of Missouri, Columbia, MO 65211, USA
| | - Ryan J. Dashek
- Research Service, Harry S Truman Memorial Veterans Medical Center, Columbia, Missouri 65212, USA
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA
| | - Grace M. Meers
- Research Service, Harry S Truman Memorial Veterans Medical Center, Columbia, Missouri 65212, USA
- Nutrition and Exercise Physiology, University of Missouri, Columbia, MO 65211, USA
| | - Vivien Jepkemoi
- Research Service, Harry S Truman Memorial Veterans Medical Center, Columbia, Missouri 65212, USA
| | - Takamune Takahashi
- Division of Nephrology and Hypertension, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | | | - Jill A. Kanaley
- Nutrition and Exercise Physiology, University of Missouri, Columbia, MO 65211, USA
| | - Frank W. Booth
- Nutrition and Exercise Physiology, University of Missouri, Columbia, MO 65211, USA
- Department of Biomedical Sciences, University of Missouri, Columbia, MO 65211, USA
| | - R. Scott Rector
- Research Service, Harry S Truman Memorial Veterans Medical Center, Columbia, Missouri 65212, USA
- Departments of Medicine-Division of Gastroenterology and Hepatology
- Nutrition and Exercise Physiology, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
22
|
Rosa-Caldwell ME, Poole KE, Seija A, Harris MP, Greene NP, Wooten JS. Exercise during weight-loss improves hepatic mitophagy. SPORTS MEDICINE AND HEALTH SCIENCE 2022; 4:183-189. [PMID: 36090917 PMCID: PMC9453692 DOI: 10.1016/j.smhs.2022.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/01/2022] [Accepted: 04/06/2022] [Indexed: 11/02/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has recently become a public health concern concurrent with the obesity crisis. Previous work has shown aberrant mitochondrial content/quality and autophagy in models of NAFLD, whereas exercise is known to improve these derangements. The purpose of this study was to examine the effect of different weight-loss modalities on hepatic mitochondrial content, autophagy and mitophagy in NAFLD. Forty-eight male C57BL/6J mice were divided into 1 of 4 groups: low fat diet (LFD, 10% fat, 18 weeks), high fat diet (HFD, 60% fat diet, 18 weeks), weight-loss by diet (D, 60% fat diet for 10 weeks then 10% fat diet for 8 weeks) or weight-loss by diet and physical activity (D/PA, 60% fat diet for 10 weeks, then 10% fat diet plus a running wheel for 8 weeks). Immunoblot data were analyzed by one-way analysis of variance (ANOVA) with significance denoted at p < 0.05. COX-IV protein contents were approximately 50% less in HFD compared to LFD. D/PA had 50% more BNIP3 compared to HFD. PINK1 content was 40% higher in D and D/PA compared to LFD. P-PARKIN/PARKIN levels were 40% lower in HFD, D, and D/PA compared to LFD. Whereas p-UbSer65 was 3-fold higher in HFD. LC3II/I ratio was 50% greater in HFD and D/PA, yet p62 protein content was 2.5 fold higher in HFD. High-fat diet causes disruptions in markers of mitochondrial quality control. Physical activity combined with diet were able to ameliorate these derangements and seemingly improve hepatic mitochondrial quality above control values.
Collapse
|
23
|
McCoin CS, Franczak E, Deng F, Pei D, Ding WX, Thyfault JP. Acute exercise rapidly activates hepatic mitophagic flux. J Appl Physiol (1985) 2022; 132:862-873. [PMID: 35142562 PMCID: PMC8934677 DOI: 10.1152/japplphysiol.00704.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 01/25/2022] [Accepted: 02/02/2022] [Indexed: 01/18/2023] Open
Abstract
Exercise is critical for improving metabolic health and putatively maintains or enhances mitochondrial quality control in metabolic tissues. Although previous work has shown that exercise elicits hepatic mitochondrial biogenesis, it is unknown if acute exercise activates hepatic mitophagy, the selective degradation of damaged or low-functioning mitochondria. We tested if an acute bout of treadmill running increased hepatic mitophagic flux both right after and 2-h postexercise in 15- to 24-wk-old C57BL/6J female mice. Acute exercise did not significantly increase markers of autophagic flux, however, mitophagic flux was activated 2-h post-treadmill running as measured by accumulation of both LC3-II and p62 in isolated mitochondria in the presence of leupeptin, an inhibitor of autophagosome degradation. Furthermore, mitochondrial-associated ubiquitin, which recruits the autophagy receptor protein p62, was also significantly increased at 2 h. Further examination via Western blot and proteomics analysis revealed that acute exercise elicits a time-dependent, dynamic activation of mitophagy pathways. Moreover, the results suggest that exercise-induced hepatic mitophagy is likely mediated by both polyubiquitination and receptor-mediated signaling pathways. Overall, we provide evidence that acute exercise activates hepatic mitophagic flux while also revealing specific receptor-mediated proteins by which exercise maintains mitochondrial quality control in the liver.NEW & NOTEWORTHY This study provides evidence that acute exercise activates hepatic mitophagic flux and mitochondrial polyubiquitination while additionally revealing specific receptor-mediated proteins by which exercise maintains mitochondrial quality control in the liver.
Collapse
Affiliation(s)
- Colin S McCoin
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, Missouri
- Center for Children's Healthy Lifestyles and Nutrition, Children's Mercy Kansas City, Kansas City, Missouri
- Kansas City Veterans Affairs Medical Center, Kansas City, Missouri
| | - Edziu Franczak
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, Missouri
| | - Fengyan Deng
- Stowers Institute for Medical Research, Kansas City, Missouri
| | - Dong Pei
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, Kansas
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - John P Thyfault
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, Missouri
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
- Center for Children's Healthy Lifestyles and Nutrition, Children's Mercy Kansas City, Kansas City, Missouri
- Kansas City Veterans Affairs Medical Center, Kansas City, Missouri
| |
Collapse
|
24
|
Cunningham RP, Moore MP, Dashek RJ, Meers GM, Takahashi T, Sheldon RD, Wheeler AA, Diaz-Arias A, Ibdah JA, Parks EJ, Thyfault JP, Rector RS. Critical Role for Hepatocyte-Specific eNOS in NAFLD and NASH. Diabetes 2021; 70:2476-2491. [PMID: 34380696 PMCID: PMC8564406 DOI: 10.2337/db20-1228] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 07/30/2021] [Indexed: 11/13/2022]
Abstract
Regulation of endothelial nitric oxide synthase (eNOS) in hepatocytes may be an important target in nonalcoholic fatty liver disease (NAFLD) development and progression to nonalcoholic steatohepatitis (NASH). In this study, we show genetic deletion and viral knockdown of hepatocyte-specific eNOS exacerbated hepatic steatosis and inflammation, decreased hepatic mitochondrial fatty acid oxidation and respiration, increased mitochondrial H2O2 emission, and impaired the hepatic mitophagic (BNIP3 and LC3II) response. Conversely, overexpressing eNOS in hepatocytes in vitro and in vivo increased hepatocyte mitochondrial respiration and attenuated Western diet-induced NASH. Moreover, patients with elevated NAFLD activity score (histology score of worsening steatosis, hepatocyte ballooning, and inflammation) exhibited reduced hepatic eNOS expression, which correlated with reduced hepatic mitochondrial fatty acid oxidation and lower hepatic protein expression of mitophagy protein BNIP3. The current study reveals an important molecular role for hepatocyte-specific eNOS as a key regulator of NAFLD/NASH susceptibility and mitochondrial quality control with direct clinical correlation to patients with NASH.
Collapse
Affiliation(s)
- Rory P Cunningham
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO
| | - Mary P Moore
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO
| | - Ryan J Dashek
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO
- Comparative Medicine Program, University of Missouri, Columbia, MO
| | - Grace M Meers
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO
| | - Takamune Takahashi
- Division of Nephrology and Hypertension, Vanderbilt University School of Medicine, Nashville, TN
| | - Ryan D Sheldon
- Metabolic and Nutritional Programming, Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, MI
| | | | | | - Jamal A Ibdah
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO
| | - Elizabeth J Parks
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO
| | - John P Thyfault
- Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS
- Kansas City VA Medical Center, Kansas City, MO
| | - R Scott Rector
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO
| |
Collapse
|
25
|
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
|
26
|
Morris EM, Noland RD, Ponte ME, Montonye ML, Christianson JA, Stanford JA, Miles JM, Hayes MR, Thyfault JP. Reduced Liver-Specific PGC1a Increases Susceptibility for Short-Term Diet-Induced Weight Gain in Male Mice. Nutrients 2021; 13:2596. [PMID: 34444756 PMCID: PMC8400659 DOI: 10.3390/nu13082596] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/21/2021] [Accepted: 07/24/2021] [Indexed: 11/18/2022] Open
Abstract
The central integration of peripheral neural signals is one mechanism by which systemic energy homeostasis is regulated. Previously, increased acute food intake following the chemical reduction of hepatic fatty acid oxidation and ATP levels was prevented by common hepatic branch vagotomy (HBV). However, possible offsite actions of the chemical compounds confound the precise role of liver energy metabolism. Herein, we used a hepatocyte PGC1a heterozygous (LPGC1a) mouse model, with associated reductions in mitochondrial fatty acid oxidation and respiratory capacity, to assess the role of liver energy metabolism in systemic energy homeostasis. LPGC1a male, but not female, mice had a 70% greater high-fat/high-sucrose (HFHS) diet-induced weight gain compared to wildtype (WT) mice (p < 0.05). The greater weight gain was associated with altered feeding behavior and lower activity energy expenditure during the HFHS diet in LPGC1a males. WT and LPGC1a mice underwent sham surgery or HBV to assess whether vagal signaling was involved in the HFHS-induced weight gain of male LPGC1a mice. HBV increased HFHS-induced weight gain (85%, p < 0.05) in male WT mice, but not LPGC1a mice. These data demonstrate a sex-specific role of reduced liver energy metabolism in acute diet-induced weight gain, and the need for a more nuanced assessment of the role of vagal signaling in short-term diet-induced weight gain.
Collapse
Affiliation(s)
- E. Matthew Morris
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (R.D.N.); (M.E.P.); (J.A.S.); (J.P.T.)
- Center for Children’s Healthy Lifestyle and Nutrition, Children’s Mercy Hospital, Kansas City, MO 64108, USA
| | - Roberto D. Noland
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (R.D.N.); (M.E.P.); (J.A.S.); (J.P.T.)
| | - Michael E. Ponte
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (R.D.N.); (M.E.P.); (J.A.S.); (J.P.T.)
| | - Michelle L. Montonye
- Department of Nutrition & Exercise Physiology, University of Missouri, Columbia, MO 65211, USA;
| | - Julie A. Christianson
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA;
| | - John A. Stanford
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (R.D.N.); (M.E.P.); (J.A.S.); (J.P.T.)
| | - John M. Miles
- Department of Internal Medicine—Division of Endocrinology and Metabolism, University of Kansas Medical Center, Kansas City, KS 66160, USA;
| | - Matthew R. Hayes
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - John P. Thyfault
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (R.D.N.); (M.E.P.); (J.A.S.); (J.P.T.)
- Center for Children’s Healthy Lifestyle and Nutrition, Children’s Mercy Hospital, Kansas City, MO 64108, USA
- Department of Internal Medicine—Division of Endocrinology and Metabolism, University of Kansas Medical Center, Kansas City, KS 66160, USA;
- Kansas City VA Medical Center-Research Service, Kansas City, MO 64128, USA
| |
Collapse
|
27
|
McKie GL, Shamshoum H, Hunt KL, Thorpe HHA, Dibe HA, Khokhar JY, Doucette CA, Wright DC. Intermittent cold exposure improves glucose homeostasis despite exacerbating diet-induced obesity in mice housed at thermoneutrality. J Physiol 2021; 600:829-845. [PMID: 34192813 DOI: 10.1113/jp281774] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 06/28/2021] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Ambient cold exposure is often regarded as a promising anti-obesity treatment in mice. However, most preclinical studies aimed at treating obesity via cold-induced thermogenesis have been confounded by subthermoneutral housing temperatures. Therefore, the ability of ambient cold to combat diet-induced obesity in mice housed under humanized thermoneutral conditions is currently unknown. Moreover, mammals such as mice are rarely exposed to chronic ambient cold without reprieve, yet mice are often subjected to experimental conditions of chronic rather than intermittent cold exposure (ICE), despite ICE being more physiologically relevant. In the present study, we provide novel evidence that thermoneutral housing uncouples the effects of ICE on glucose and energy homeostasis suggesting that ICE, despite improving glucose tolerance, is not an effective obesity treatment when mice are housed under humanized thermoneutral conditions. ABSTRACT The present study examines whether a physiologically relevant model of ambient cold exposure, intermittent cold exposure (ICE), could ameliorate the metabolic impairments of diet-induced obesity in male and female mice housed under humanized thermoneutral conditions. Male and female C57BL/6J mice housed at thermoneutrality (29°C) were fed a low-fat diet or high-fat diet for 6 weeks before being weight matched into groups that remained unperturbed or underwent ICE for 4 weeks (4°C for 60 min day-1 ; 5 days week-1 ) when being maintained on their respective diets. ICE induced rapid and persistent hyperphagia exacerbating rather than attenuating high-fat diet-induced obesity over time. These ICE-induced increases in adiposity were found to be energy intake-dependent via pair-feeding. Despite exacerbating high-fat diet-induced obesity, ICE improved glucose tolerance, independent of diet, in a sex-specific manner. The effects of ICE on glucose tolerance were not attributed to improvements in whole-body insulin tolerance, tissue specific insulin action, nor differences in markers of hepatic insulin clearance or pancreatic beta cell proliferation. Instead, ICE increased serum concentrations of insulin and C-peptide in response to glucose, suggesting that ICE may improve glucose tolerance by potentiating pancreatic glucose-stimulated insulin secretion. These data suggest that ICE, despite improving glucose tolerance, is not an effective obesity treatment in mice housed under humanized conditions.
Collapse
Affiliation(s)
- Greg L McKie
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Hesham Shamshoum
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Kristin L Hunt
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB, Canada
| | - Hayley H A Thorpe
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, Canada
| | - Hana A Dibe
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Jibran Y Khokhar
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, Canada
| | - Christine A Doucette
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB, Canada
| | - David C Wright
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
28
|
McKie GL, Wright DC. The confounding effects of sub-thermoneutral housing temperatures on aerobic exercise-induced adaptations in mouse subcutaneous white adipose tissue. Biol Lett 2021; 17:20210171. [PMID: 34186002 DOI: 10.1098/rsbl.2021.0171] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Mice are the most commonly used model organism for human biology, and failure to acknowledge fundamental differences in thermal biology between these species has confounded the study of adipose tissue metabolism in mice and its translational relevance to humans. Here, using exercise biochemistry as an example, we highlight the subtle yet detrimental effects sub-thermoneutral housing temperatures can have on the study of adipose tissue metabolism in mice. We encourage academics and publishers to consider ambient housing temperature as a key determinant in the methodological conception and reporting of all research on rodent white adipose tissue metabolism.
Collapse
Affiliation(s)
- Greg L McKie
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Road East, Guelph, Ontario, Canada N1G 2W1
| | - David C Wright
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Road East, Guelph, Ontario, Canada N1G 2W1
| |
Collapse
|
29
|
Dankel SN, Bjørndal B, Lindquist C, Grinna ML, Rossmann CR, Bohov P, Nygård O, Hallström S, Strand E, Berge RK. Hepatic Energy Metabolism Underlying Differential Lipidomic Responses to High-Carbohydrate and High-Fat Diets in Male Wistar Rats. J Nutr 2021; 151:2610-2621. [PMID: 34132338 PMCID: PMC8417924 DOI: 10.1093/jn/nxab178] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/17/2021] [Accepted: 05/11/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Low-carbohydrate diets are suggested to exert metabolic benefits by reducing circulating triacylglycerol (TG) concentrations, possibly by enhancing mitochondrial activity. OBJECTIVE We aimed to elucidate mechanisms by which dietary carbohydrate and fat differentially affect hepatic and circulating TG, and how these mechanisms relate to fatty acid composition. METHODS Six-week-old, ∼300 g male Wistar rats were fed a high-carbohydrate, low-fat [HC; 61.3% of energy (E%) carbohydrate] or a low-carbohydrate, high-fat (HF; 63.5 E% fat) diet for 4 wk. Parameters of lipid metabolism and mitochondrial function were measured in plasma and liver, with fatty acid composition (GC), high-energy phosphates (HPLC), carnitine metabolites (HPLC-MS/MS), and hepatic gene expression (qPCR) as main outcomes. RESULTS In HC-fed rats, plasma TG was double and hepatic TG 27% of that in HF-fed rats. The proportion of oleic acid (18:1n-9) was 60% higher after HF vs. HC feeding while the proportion of palmitoleic acid (16:1n-7) and vaccenic acid (18:1n-7), and estimated activities of stearoyl-CoA desaturase, SCD-16 (16:1n-7/16:0), and de novo lipogenesis (16:0/18:2n-6) were 1.5-7.5-fold in HC vs. HF-fed rats. Accordingly, hepatic expression of fatty acid synthase (Fasn) and acetyl-CoA carboxylase (Acaca/Acc) was strongly upregulated after HC feeding, accompanied with 8-fold higher FAS activity and doubled ACC activity. There were no differences in expression of liver-specific biomarkers of mitochondrial biogenesis and activity (Cytc, Tfam, Cpt1, Cpt2, Ucp2, Hmgcs2); concentrations of ATP, AMP, and energy charge; plasma carnitine/acylcarnitine metabolites; or peroxisomal fatty acid oxidation. CONCLUSIONS In male Wistar rats, dietary carbohydrate was converted into specific fatty acids via hepatic lipogenesis, contributing to higher plasma TG and total fatty acids compared with high-fat feeding. In contrast, the high-fat, low-carbohydrate feeding increased hepatic fatty acid content, without affecting hepatic mitochondrial fatty acid oxidation.
Collapse
Affiliation(s)
| | - Bodil Bjørndal
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Carine Lindquist
- Department of Clinical Science, University of Bergen, Bergen, Norway,Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Mari L Grinna
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | | | - Pavol Bohov
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Ottar Nygård
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway,Department of Clinical Science, University of Bergen, Bergen, Norway,Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Seth Hallström
- Division of Physiological Chemistry, Medical University of Graz, Graz, Austria
| | - Elin Strand
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | | |
Collapse
|
30
|
Fuller KNZ, McCoin CS, Von Schulze AT, Houchen CJ, Choi MA, Thyfault JP. Estradiol treatment or modest exercise improves hepatic health and mitochondrial outcomes in female mice following ovariectomy. Am J Physiol Endocrinol Metab 2021; 320:E1020-E1031. [PMID: 33870713 PMCID: PMC8285602 DOI: 10.1152/ajpendo.00013.2021] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/16/2021] [Accepted: 04/13/2021] [Indexed: 02/06/2023]
Abstract
We recently reported that compared with males, female mice have increased hepatic mitochondrial respiratory capacity and are protected against high-fat diet-induced steatosis. Here, we sought to determine the role of estrogen in hepatic mitochondrial function, steatosis, and bile acid metabolism in female mice and investigate potential benefits of exercise in the absence or presence of estrogen via ovariectomy (OVX). Female C57BL mice (n = 6 per group) were randomly assigned to sham surgery (sham), ovariectomy (OVX), or OVX plus estradiol replacement therapy (OVX + Est). Half of the mice in each treatment group were sedentary (SED) or had access to voluntary wheel running (VWR). All mice were fed a high-fat diet (HFD) and were housed at thermoneutral temperatures. We assessed isolated hepatic mitochondrial respiratory capacity using the Oroboros O2k with both pyruvate and palmitoylcarnitine as substrates. As expected, OVX mice presented with greater hepatic steatosis, weight gain, and fat mass gain compared with sham and OVX + Est animals. Hepatic mitochondrial coupling (basal/state 3 respiration) with pyruvate was impaired following OVX, but both VWR and estradiol treatment rescued coupling to levels greater than or equal to sham animals. Estradiol and exercise also had different effects on liver electron transport chain protein expression depending on OVX status. Markers of bile acid metabolism and excretion were also impaired by ovariectomy but rescued with estradiol add-back. Together our data suggest that estrogen depletion impairs hepatic mitochondrial function and liver health, and that estradiol replacement and modest exercise can aid in rescuing this phenotype.NEW & NOTEWORTHY OVX induces hepatic steatosis in sedentary mice which can be prevented by modest physical activity (VWR) and/or estradiol treatment. Estrogen impacts hepatic mitochondrial coupling in a substrate-specific manner. OVX mice have impaired fecal bile acid excretion, which was rescued with estradiol treatment.
Collapse
Affiliation(s)
- Kelly N Z Fuller
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
- Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, Kansas
| | - Colin S McCoin
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
- Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, Kansas
- Center for Children's Healthy Lifestyles and Nutrition, Kansas City, Missouri
| | - Alex T Von Schulze
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Claire J Houchen
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Michael A Choi
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - John P Thyfault
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
- Department of Internal Medicine, Division of Endocrinology and Metabolism, University of Kansas Medical Center, Kansas City, Kansas
- Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, Kansas
- Center for Children's Healthy Lifestyles and Nutrition, Kansas City, Missouri
| |
Collapse
|
31
|
Longo M, Meroni M, Paolini E, Macchi C, Dongiovanni P. Mitochondrial dynamics and nonalcoholic fatty liver disease (NAFLD): new perspectives for a fairy-tale ending? Metabolism 2021; 117:154708. [PMID: 33444607 DOI: 10.1016/j.metabol.2021.154708] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 01/05/2021] [Accepted: 01/07/2021] [Indexed: 12/12/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) includes a broad spectrum of liver dysfunctions and it is predicted to become the primary cause of liver failure and hepatocellular carcinoma. Mitochondria are highly dynamic organelles involved in multiple metabolic/bioenergetic pathways in the liver. Emerging evidence outlined that hepatic mitochondria adapt in number and functionality in response to external cues, as high caloric intake and obesity, by modulating mitochondrial biogenesis, and maladaptive mitochondrial response has been described from the early stages of NAFLD. Indeed, mitochondrial plasticity is lost in progressive NAFLD and these organelles may assume an aberrant phenotype to drive or contribute to hepatocarcinogenesis. Severe alimentary regimen and physical exercise represent the cornerstone for NAFLD care, although the low patients' compliance is urging towards the discovery of novel pharmacological treatments. Mitochondrial-targeted drugs aimed to recover mitochondrial lifecycle and to modulate oxidative stress are becoming attractive molecules to be potentially introduced for NAFLD management. Although the path guiding the switch from bench to bedside remains tortuous, the study of mitochondrial dynamics is providing intriguing perspectives for future NAFLD healthcare.
Collapse
Affiliation(s)
- Miriam Longo
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Pad. Granelli, via F Sforza 35, 20122 Milan, Italy; Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milano, Italy
| | - Marica Meroni
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Pad. Granelli, via F Sforza 35, 20122 Milan, Italy; Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20122 Milano, Italy
| | - Erika Paolini
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Pad. Granelli, via F Sforza 35, 20122 Milan, Italy; Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milano, Italy
| | - Chiara Macchi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milano, Italy
| | - Paola Dongiovanni
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Pad. Granelli, via F Sforza 35, 20122 Milan, Italy.
| |
Collapse
|
32
|
Von Schulze AT, Deng F, Fuller KNZ, Franczak E, Miller J, Allen J, McCoin CS, Shankar K, Ding WX, Thyfault JP, Geiger PC. Heat Treatment Improves Hepatic Mitochondrial Respiratory Efficiency via Mitochondrial Remodeling. FUNCTION (OXFORD, ENGLAND) 2021; 2:zqab001. [PMID: 33629069 PMCID: PMC7886620 DOI: 10.1093/function/zqab001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/24/2020] [Accepted: 12/30/2020] [Indexed: 01/06/2023]
Abstract
Nonacholic fatty liver disease, or hepatic steatosis, is the most common liver disorder affecting the western world and currently has no pharmacologic cure. Thus, many investigations have focused on alternative strategies to treat or prevent hepatic steatosis. Our laboratory has shown that chronic heat treatment (HT) mitigates glucose intolerance, insulin resistance, and hepatic steatosis in rodent models of obesity. Here, we investigate the direct bioenergetic mechanism(s) surrounding the metabolic effects of HT on hepatic mitochondria. Utilizing mitochondrial proteomics and respiratory function assays, we show that one bout of acute HT (42°C for 20 min) in male C57Bl/6J mice (n = 6/group) triggers a hepatic mitochondrial heat shock response resulting in acute reductions in respiratory capacity, degradation of key mitochondrial enzymes, and induction of mitophagy via mitochondrial ubiquitination. We also show that chronic bouts of HT and recurrent activation of the heat shock response enhances mitochondrial quality and respiratory function via compensatory adaptations in mitochondrial organization, gene expression, and transport even during 4 weeks of high-fat feeding (n = 6/group). Finally, utilizing a liver-specific heat shock protein 72 (HSP72) knockout model, we are the first to show that HSP72, a protein putatively driving the HT metabolic response, does not play a significant role in the hepatic mitochondrial adaptation to acute or chronic HT. However, HSP72 is required for the reductions in blood glucose observed with chronic HT. Our data are the first to suggest that chronic HT (1) improves hepatic mitochondrial respiratory efficiency via mitochondrial remodeling and (2) reduces blood glucose in a hepatic HSP72-dependent manner.
Collapse
Affiliation(s)
- Alex T Von Schulze
- Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Fengyan Deng
- Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Kelly N Z Fuller
- Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Edziu Franczak
- Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Josh Miller
- Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Julie Allen
- Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Colin S McCoin
- Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Kartik Shankar
- Pediatrics, Section of Nutrition, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Wen-Xing Ding
- Pharmacology, Toxicology & Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160 USA
| | - John P Thyfault
- Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Paige C Geiger
- Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, KS 66160, USA,Address correspondence to P.C.G. (e-mail: )
| |
Collapse
|
33
|
Fuller KNZ, Thyfault JP. Barriers in translating preclinical rodent exercise metabolism findings to human health. J Appl Physiol (1985) 2021; 130:182-192. [PMID: 33180643 PMCID: PMC7944931 DOI: 10.1152/japplphysiol.00683.2020] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/21/2020] [Accepted: 11/10/2020] [Indexed: 01/03/2023] Open
Abstract
Physical inactivity and low aerobic capacity are primary drivers of chronic disease pathophysiology and are independently associated with all-cause mortality. Conversely, increased physical activity and exercise are central to metabolic disease prevention and longevity. Although these relationships are well characterized in the literature, what remains incompletely understood are the mechanisms by which physical activity/exercise prevents disease. Given methodological constraints of clinical research, investigators must often rely on preclinical rodent models to investigate these potential underlying mechanisms. However, there are several key barriers to applying exercise metabolism findings from rodent models to human health. These barriers include housing temperature, nutrient metabolism, exercise modality, exercise testing, and sex differences. Increased awareness and understanding of these barriers will enhance the ability to impact human health through more appropriate experimental design and interpretation of data within the context of these factors.
Collapse
Affiliation(s)
- Kelly N Z Fuller
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - John P Thyfault
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
- Research Service Kansas City Veterans Affairs Medical Center, Kansas City, Kansas
- Center for Children's Healthy Lifestyles and Nutrition, Kansas City, Missouri
| |
Collapse
|
34
|
Ren XL, Han P, Meng Y. Aflatoxin B1-Induced COX-2 Expression Promotes Mitophagy and Contributes to Lipid Accumulation in Hepatocytes In Vitro and In Vivo. Int J Toxicol 2020; 39:594-604. [PMID: 32687719 DOI: 10.1177/1091581820939081] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
AIM Aflatoxin B1 (AFB1) is hepatotoxic. Numerous studies have shown that mitochondria play an essential role in AFB1-induced steatosis. However, the mechanisms of AFB1-induced steatosis via mitochondria are still obscure. The present study aimed to confirm that AFB1 causes hepatocyte steatosis regulated by cyclooxygenase-2 (COX-2)-induced mitophagy, both in vivo and in vitro. METHODS Adult male C57BL/6 mice were randomly divided into control group with the same volume of peanut oil and exposure group administered 0.6 mg/kg AFB1 once in 2 days for 1 month. HepG2 and Cas9-PTGS2 cells were treated with 5 μM AFB1 for 48 hours. Then, various indicators were evaluated. RESULTS Aflatoxin B1 causes liver injury and steatosis with increased alanine aminotransferase, aspartate aminotransferase, total cholesterol, total triglyceride levels in vivo and in vitro, and elevated lipid droplets in HepG2 cells. Cyclooxygenase-2 and mitophagy pathway were induced by AFB1 in both liver tissues and cultured HepG2 cells. Further studies have shown that knockout of COX-2 with the CRISPR/Cas9 system inhibited the AFB1-induced mitophagy and steatosis in HepG2 cells. Also, the inhibition of PTEN-induced putative kinase with RNA interference attenuated the AFB1-induced steatosis. CONCLUSIONS The results of the current study suggested that AFB1 increases the expression of COX-2, which, in turn, elevates the level of mitophagy, thereby disrupting the normal mitochondrial lipid metabolism and causing steatosis. Thus, this study implies that COX-2 may be a potential target for therapy against AFB1-induced steatosis.
Collapse
Affiliation(s)
- Xin-Lu Ren
- Queen Mary College of Nanchang University, Nanchang, China
| | - Peiyu Han
- Wuxi School of Medicine, 66374Jiangnan University, Wuxi, China
| | - Yiteng Meng
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| |
Collapse
|
35
|
Fuller KNZ, McCoin CS, Allen J, Bell-Glenn S, Koestler DC, Dorn GW, Thyfault JP. Sex and BNIP3 genotype, rather than acute lipid injection, modulate hepatic mitochondrial function and steatosis risk in mice. J Appl Physiol (1985) 2020; 128:1251-1261. [PMID: 32240015 PMCID: PMC7272752 DOI: 10.1152/japplphysiol.00035.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/10/2020] [Accepted: 03/27/2020] [Indexed: 12/23/2022] Open
Abstract
Both lipid oversupply and poor mitochondrial function (low respiration and elevated H2O2 emission) have been implicated in the development of hepatic steatosis and liver injury. Mitophagy, the targeted degradation of low-functioning mitochondria, is critical for maintaining mitochondrial quality control. Here, we used intralipid injection combined with acute (4 day) and chronic (4-7wk) high-fat diets (HFD) to examine whether hepatic mitochondrial respiration would decrease and H2O2 emission would increase with lipid overload. We tested these effects in male and female wild type (WT) mice and mice null for a critical mediator of mitophagy, BCL-2/adenovirus EIB 19-kDa interacting protein knockout (BNIP3 KO) housed at thermoneutral temperatures. Intralipid injection was successful in elevating serum triglycerides and nonesterified fatty acids but had no impact on hepatic mitochondrial respiratory function or H2O2 emission. However, female mice had greater mitochondrial respiration on the acute HFD and lower H2O2 emission across both HFD durations and were protected against hepatic steatosis. Unexpectedly, BNIP3 KO animals had greater hepatic mitochondrial respiration, better coupled respiration, and increased electron chain protein content after the 4-day HFD, compared with WT animals. Altogether, these data suggest that acute lipid overload delivered by a single intralipid bolus does not alter hepatic mitochondrial outcomes, but rather sex and genotype profoundly impact hepatic mitochondrial respiration and H2O2 emission.NEW & NOTEWORTHY This is the first study focusing on hepatic mitochondrial respiratory outcomes in response to lipid overload via a high-fat diet (HFD) combined with intralipid injection. Novel findings include no effect of intralipid injection on mitochondrial outcomes of interest despite increased circulating lipid concentrations. However, we report pronounced differences in hepatic mitochondrial respiration, complex protein expression, and H2O2 production by sex and BCL-2/adenovirus EIB 19-kDa interacting protein (BNIP3) genotype. Specifically, female mice had lower H2O2 emission globally and on an acute HFD, females had greater hepatic mitochondrial respiration than males while BNIP3 knockout (KO) animals had greater mitochondrial coupling and complex protein expression than wild-type (WT) animals.
Collapse
Affiliation(s)
- Kelly N Z Fuller
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Colin S McCoin
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
- Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, Kansas
- Center for Children's Healthy Lifestyles and Nutrition, Kansas City, Missouri
| | - Julie Allen
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
- Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, Kansas
| | - Shelby Bell-Glenn
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, Kansas
| | - Devin C Koestler
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, Kansas
| | - Gerald W Dorn
- Center for Pharmacogenomics, Department of Internal Medicine, Washington University, School of Medicine, St. Louis, Missouri
| | - John P Thyfault
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
- Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, Kansas
- Center for Children's Healthy Lifestyles and Nutrition, Kansas City, Missouri
| |
Collapse
|
36
|
Salvoza NC, Giraudi PJ, Tiribelli C, Rosso N. Sex differences in non-alcoholic fatty liver disease: hints for future management of the disease. EXPLORATION OF MEDICINE 2020; 1:51-74. [DOI: 10.37349/emed.2020.00005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 04/16/2020] [Indexed: 01/04/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) remains a major cause of chronic liver disease worldwide. Despite extensive studies, the heterogeneity of the risk factors as well as different disease mechanisms complicate the goals toward effective diagnosis and management. Recently, it has been shown that sex differences play a role in the prevalence and progression of NAFLD. In vitro, in vivo, and clinical studies revealed that the lower prevalence of NAFLD in premenopausal as compared to postmenopausal women and men is mainly due to the protective effects of estrogen and body fat distribution. It has been also described that males and females present differential pathogenic features in terms of biochemical profiles and histological characteristics. However, the exact molecular mechanisms for the gender differences that exist in the pathogenesis of NAFLD are still elusive. Lipogenesis, oxidative stress, and inflammation play a key role in the progression of NAFLD. For NAFLD, only a few studies characterized these mechanisms at the molecular level. Therefore, we aim to review the reported differential molecular mechanisms that trigger such different pathogenesis in both sexes. Differences in lipid metabolism, glucose homeostasis, oxidative stress, inflammation, and fibrosis were discussed based on the evidence reported in recent publications. In conclusion, with this review, we hope to provide a new perspective for the development of future practice guidelines as well as a new avenue for the management of the disease.
Collapse
Affiliation(s)
- Noel C. Salvoza
- Fondazione Italiana Fegato ONLUS, Area Science Park Basovizza SS14 km 163.5, 34149 Trieste, Italy; Philippine Council for Health Research and Development, DOST Compound, Bicutan Taguig City 1631, Philippines
| | - Pablo J. Giraudi
- Fondazione Italiana Fegato ONLUS, Area Science Park Basovizza SS14 km 163.5, 34149 Trieste, Italy
| | - Claudio Tiribelli
- Fondazione Italiana Fegato ONLUS, Area Science Park Basovizza SS14 km 163.5, 34149 Trieste, Italy
| | - Natalia Rosso
- Fondazione Italiana Fegato ONLUS, Area Science Park Basovizza SS14 km 163.5, 34149 Trieste, Italy
| |
Collapse
|
37
|
Lonardo A, Suzuki A. Sexual Dimorphism of NAFLD in Adults. Focus on Clinical Aspects and Implications for Practice and Translational Research. J Clin Med 2020; 9:jcm9051278. [PMID: 32354182 PMCID: PMC7288212 DOI: 10.3390/jcm9051278] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) embraces the clinico-pathological consequences of hepatic lipotoxicity and is a major public health problem globally. Sexual dimorphism is a definite feature of most human diseases but, under this aspect, NAFLD lags behind other medical fields. Here, we aim at summarizing and critically discussing the most prominent sex differences and gaps in NAFLD in humans, with emphasis on those aspects which are relevant for clinical practice and translational research. Sexual dimorphism of NAFLD is covered with references to the following areas: disease prevalence and risk factors, pathophysiology, comorbidities, natural course and complications. Finally, we also discuss selected gender differences and whether sex-specific lifestyle changes should be adopted to contrast NAFLD in men and women.
Collapse
Affiliation(s)
- Amedeo Lonardo
- Operating Unit Metabolic Syndrome, Azienda Ospedaliero-Universitaria di Modena, Ospedale Civile di Baggiovara, 41126 Baggiovara MO, Italy
- Correspondence:
| | - Ayako Suzuki
- Division of Gastroenterology, Durham VA Medical Center and Duke University Medical Center, Durham, NC 27705, USA;
| |
Collapse
|
38
|
Léveillé M, Estall JL. Mitochondrial Dysfunction in the Transition from NASH to HCC. Metabolites 2019; 9:E233. [PMID: 31623280 PMCID: PMC6836234 DOI: 10.3390/metabo9100233] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 09/26/2019] [Accepted: 10/11/2019] [Indexed: 02/06/2023] Open
Abstract
The liver constantly adapts to meet energy requirements of the whole body. Despite its remarkable adaptative capacity, prolonged exposure of liver cells to harmful environmental cues (such as diets rich in fat, sugar, and cholesterol) results in the development of chronic liver diseases (including non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH)) that can progress to hepatocellular carcinoma (HCC). The pathogenesis of these diseases is extremely complex, multifactorial, and poorly understood. Emerging evidence suggests that mitochondrial dysfunction or maladaptation contributes to detrimental effects on hepatocyte bioenergetics, reactive oxygen species (ROS) homeostasis, endoplasmic reticulum (ER) stress, inflammation, and cell death leading to NASH and HCC. The present review highlights the potential contribution of altered mitochondria function to NASH-related HCC and discusses how agents targeting this organelle could provide interesting treatment strategies for these diseases.
Collapse
Affiliation(s)
- Mélissa Léveillé
- Institut de Recherches Cliniques de Montréal (IRCM), Montreal, Quebec, QC H2W 1R7, Canada.
- Faculty of Medicine, University of Montreal, Montreal, Quebec, QC H3G 2M1, Canada.
| | - Jennifer L Estall
- Institut de Recherches Cliniques de Montréal (IRCM), Montreal, Quebec, QC H2W 1R7, Canada.
- Faculty of Medicine, University of Montreal, Montreal, Quebec, QC H3G 2M1, Canada.
- Division of Experimental Medicine, McGill University, Montreal, Quebec, QC H4A 3J1, Canada.
| |
Collapse
|