1
|
Skawratananond S, Xiong DX, Zhang C, Tonk S, Pinili A, Delacruz B, Pham P, Smith SC, Navab R, Reddy PH. Mitophagy in Alzheimer's disease and other metabolic disorders: A focus on mitochondrial-targeted therapeutics. Ageing Res Rev 2025; 108:102732. [PMID: 40122398 DOI: 10.1016/j.arr.2025.102732] [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: 11/15/2024] [Revised: 02/19/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
Mitochondria, as central regulators of cellular processes such as energy production, apoptosis, and metabolic homeostasis, are essential to cellular function and health. The maintenance of mitochondrial integrity, especially through mitophagy-the selective removal of impaired mitochondria-is crucial for cellular homeostasis. Dysregulation of mitochondrial function, dynamics, and biogenesis is linked to neurodegenerative and metabolic diseases, notably Alzheimer's disease (AD), which is increasingly recognized as a metabolic disorder due to its shared pathophysiologic features: insulin resistance, oxidative stress, and chronic inflammation. In this review, we highlight recent advancements in pharmacological interventions, focusing on agents that modulate mitophagy, mitochondrial uncouplers that reduce oxidative phosphorylation, compounds that directly scavenge reactive oxygen species to alleviate oxidative stress, and molecules that ameliorate amyloid beta plaque accumulation and phosphorylated tau pathology. Additionally, we explore dietary and lifestyle interventions-MIND and ketogenic diets, caloric restriction, physical activity, hormone modulation, and stress management-that complement pharmacological approaches and support mitochondrial health. Our review underscores mitochondria's central role in the pathogenesis and potential treatment of neurodegenerative and metabolic diseases, particularly AD. By advocating for an integrated therapeutic model that combines pharmacological and lifestyle interventions, we propose a comprehensive approach aimed at mitigating mitochondrial dysfunction and improving clinical outcomes in these complex, interrelated diseases.
Collapse
Affiliation(s)
- Shadt Skawratananond
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States.
| | - Daniel X Xiong
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX 78712, United States.
| | - Charlie Zhang
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Honors College, Texas Tech University, Lubbock, TX 79401, United States; Department of Biology, Texas Tech University, Lubbock, TX 79401, USA, Texas Tech University, Lubbock, TX 79401, United States.
| | - Sahil Tonk
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States.
| | - Aljon Pinili
- Honors College, Texas Tech University, Lubbock, TX 79401, United States; Department of Biology, Texas Tech University, Lubbock, TX 79401, USA, Texas Tech University, Lubbock, TX 79401, United States.
| | - Brad Delacruz
- Honors College, Texas Tech University, Lubbock, TX 79401, United States; Department of Biology, Texas Tech University, Lubbock, TX 79401, USA, Texas Tech University, Lubbock, TX 79401, United States.
| | - Patrick Pham
- Honors College, Texas Tech University, Lubbock, TX 79401, United States; Department of Biology, Texas Tech University, Lubbock, TX 79401, USA, Texas Tech University, Lubbock, TX 79401, United States.
| | - Shane C Smith
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States.
| | - Rahul Navab
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Department of Internal Medicine, PES Institute of Medical Sciences and Research, Kuppam, India.
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Nutritional Sciences Department, College Human Sciences, Texas Tech University, Lubbock, TX 79409, United States; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
2
|
Liu Y, Hu Y, Shan ZL. Mitochondrial DNA release mediates metabolic-associated steatohepatitis via activation of inflammatory pathways. Shijie Huaren Xiaohua Zazhi 2025; 33:344-360. [DOI: 10.11569/wcjd.v33.i5.344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/25/2025] [Accepted: 05/19/2025] [Indexed: 05/28/2025] Open
Affiliation(s)
- Ying Liu
- Gannan Institute of Medical Innovation and Translational Medicine, Gannan Medical University, Ganzhou 431000, Jiangxi Province, China
| | - Yang Hu
- Gannan Institute of Medical Innovation and Translational Medicine, Gannan Medical University, Ganzhou 431000, Jiangxi Province, China
| | - Zhao-Liang Shan
- Gannan Institute of Medical Innovation and Translational Medicine, Gannan Medical University, Ganzhou 431000, Jiangxi Province, China
| |
Collapse
|
3
|
Ponte ME, Prom JC, Newcomb MA, Jordan AB, Comfort LL, Hu J, Puchalska P, Koestler DC, Geisler CE, Hayes MR, Morris EM. Reduced liver mitochondrial energy metabolism impairs food intake regulation following gastric preloads and fasting. Mol Metab 2025; 97:102167. [PMID: 40368160 DOI: 10.1016/j.molmet.2025.102167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2025] [Revised: 05/06/2025] [Accepted: 05/07/2025] [Indexed: 05/16/2025] Open
Abstract
OBJECTIVE The capacity of the liver to serve as a peripheral sensor in the regulation of food intake has been debated for over half a century. The anatomical position and physiological roles of the liver suggest it is a prime candidate to serve as an interoceptive sensor of peripheral tissue and systemic energy state. Importantly, maintenance of liver ATP levels and within-meal food intake inhibition is impaired in human subjects with obesity and obese pre-clinical models. Previously, we have shown decreased hepatic mitochondrial energy metabolism (i.e., oxidative metabolism & ADP-dependent respiration) in male liver-specific, heterozygous PGC1a mice results in increased short-term diet-induced weight gain with increased within meal food intake. Herein, we tested the hypothesis that decreased liver mitochondrial energy metabolism impairs meal termination following nutrient oral pre-loads. METHODS Liver mitochondrial respiratory response to changes in ΔGATP and adenine nucleotide concentration following fasting were examined in male liver-specific, heterozygous PGC1a mice. Further, food intake and feeding behavior during basal conditions, following nutrient oral pre-loads, and following fasting were investigated. RESULTS We observed male liver-specific, heterozygous PGC1a mice have reduced mitochondrial response to changes in ΔGATP and tissue ATP following fasting. These impairments in liver energy state are associated with larger and longer meals during chow feeding, impaired dose-dependent food intake inhibition in response to mixed and individual nutrient oral pre-loads, and greater acute fasting-induced food intake. CONCLUSIONS These data support previous work proposing liver-mediated food intake regulation through modulation of peripheral satiation signals.
Collapse
Affiliation(s)
- Michael E Ponte
- Depatment of Cell Biology & Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - John C Prom
- Depatment of Cell Biology & Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Mallory A Newcomb
- Depatment of Cell Biology & Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Annabelle B Jordan
- Depatment of Cell Biology & Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Lucas L Comfort
- Depatment of Cell Biology & Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Jiayin Hu
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - Patrycja Puchalska
- Division of Molecular Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Devin C Koestler
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Caroline E Geisler
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA; Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY, USA
| | - Matthew R Hayes
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - E Matthew Morris
- Depatment of Cell Biology & Physiology, University of Kansas Medical Center, Kansas City, KS, USA; Center for Children's Healthy Lifestyle and Nutrition, Children's Mercy Hospital, Kansas City, MO, USA; University of Kansas Diabetes Institute, Kansas City, KS, USA.
| |
Collapse
|
4
|
Shahmohammadi A, Mirahmadi SMS, Rousta AM, Baluchnejadmojarad T, Roghani M. Protective effect of sinomenine against CCl4-induced acute liver injury through regulation of mitochondrial biogenesis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:2815-2822. [PMID: 39283527 DOI: 10.1007/s00210-024-03448-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 09/09/2024] [Indexed: 03/19/2025]
Abstract
Carbon tetrachloride (CCl4)-provoked acute liver injury (ALI) is typified by intensified apoptotic, inflammatory, and oxidative changes besides mitochondrial dysfunction. Sinomenine is an active constituent in the medicinal plant Sinomenium acutum. The main objective of this study was to determine sinomenine-induced hepatoprotection following CCl4 challenge with an emphasis on unraveling the contribution of mitochondrial biogenesis-related factors. To induce ALI, CCl4 was injected i.p. and sinomenine was orally administered at 10, 25, and 50 mg/kg. Serum factors in relation to liver dysfunction were measured in addition to hepatic analysis of apoptotic, mitochondrial biogenesis, oxidative, and inflammatory parameters. Sinomenine pretreatment significantly lowered ALT and AST, MDA, IL-6, apoptosis intensity, and TNF-α and restored mitochondrial biogenesis besides enhancement of SOD, sirtuin-1, and AMPK. Sinomenine also conferred hepatoprotective impact, as was apparent by lower pathologic changes. These effects were accompanied by changes in gene expression for AMPK/sirtuin-1/PGC-1α/PPARγ. The current study showed sinomenine hepatoprotective impact in CCl4-induced ALI that is associated with its regulation of mitochondrial biogenesis and parallel enhancement of AMPK/sirtuin-1.
Collapse
Affiliation(s)
| | | | | | | | - Mehrdad Roghani
- Neurophysiology Research Center, Shahed University, Tehran, Iran
| |
Collapse
|
5
|
Guerbette T, Ciesielski V, Brien M, Catheline D, Viel R, Bostoën M, Perrin JB, Burel A, Janvier R, Rioux V, Lan A, Boudry G. Bioenergetic adaptations of small intestinal epithelial cells reduce cell differentiation enhancing intestinal permeability in obese mice. Mol Metab 2025; 92:102098. [PMID: 39814101 PMCID: PMC11795564 DOI: 10.1016/j.molmet.2025.102098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 01/08/2025] [Indexed: 01/18/2025] Open
Abstract
OBJECTIVE Obesity and overweight are associated with low-grade inflammation induced by adipose tissue expansion and perpetuated by altered intestinal homeostasis, including increased epithelial permeability. Intestinal epithelium functions are supported by intestinal epithelial cells (IEC) mitochondria function. However, diet-induced obesity (DIO) may impair mitochondrial activity of IEC and consequently, intestinal homeostasis. The aim of the project was to determine whether DIO alters the mitochondrial function of IEC, and what are the consequences on intestinal homeostasis. METHODS C57Bl/6J mice were fed a control diet for 22 weeks or a high fat diet (58 kcal% fat). Bioenergetic adaptations of IEC were evaluated on isolated crypts and villi from mouse jejunum. To determine the link between mitochondrial function and alterations of intestinal homeostasis in response to lipid overload, we used the jejunal epithelial cell line IPEC-J2 in vitro and mouse jejunum organoids. RESULTS Here, we report that DIO in mice induced lipid metabolism adaptations favoring lipid storage in IEC together with reduced number, altered dynamics and diminished oxidative phosphorylation activity of IEC mitochondria. Using the IPEC-J2 cell line, we showed that IEC lipid metabolism and oxidative stress machinery adaptations preceded mitochondrial bioenergetic ones. Moreover, we unraveled the intricate link between IEC energetic status and proliferation / differentiation balance since enhancing mitochondrial function with the AMPK activator AICAR in jejunal organoids reduced proliferation and initiated IEC differentiation and conversely. We confirmed that the reduced IEC mitochondrial function observed in DIO mice was associated with increased proliferation and reduced differentiation, promoting expression of the permissive Cldn2 in the jejunal epithelium of DIO mice. CONCLUSIONS Our study provides new insights into metabolic adaptations of IEC in obesity by revealing that excess lipid intake diminishes mitochondrial number in IEC, reducing IEC differentiation that contribute to increased epithelial permeability.
Collapse
Affiliation(s)
| | - Vincent Ciesielski
- Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, France; Institut Agro Rennes Angers, Rennes, France
| | - Manon Brien
- Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, France
| | - Daniel Catheline
- Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, France; Institut Agro Rennes Angers, Rennes, France
| | - Roselyne Viel
- Univ Rennes, CNRS, Inserm, Biosit UAR 3480 US_S 018, France-BioImaging (ANR-10-INBS-04), plateforme H2P2, Rennes, France
| | - Mégane Bostoën
- Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, France
| | | | - Agnès Burel
- Plateforme MRic, UMS 3480 BIOSIT, Rennes, France
| | - Régis Janvier
- Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, France
| | - Vincent Rioux
- Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, France; Institut Agro Rennes Angers, Rennes, France
| | - Annaïg Lan
- Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, France; Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, 91120, Palaiseau, France
| | - Gaëlle Boudry
- Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, France.
| |
Collapse
|
6
|
Ponte ME, Prom JC, Newcomb MA, Jordan AB, Comfort LL, Hu J, Puchalska P, Geisler CE, Hayes MR, Morris EM. Reduced Liver Mitochondrial Energy Metabolism Impairs Food Intake Regulation Following Gastric Preloads and Fasting. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.24.620086. [PMID: 39554188 PMCID: PMC11565831 DOI: 10.1101/2024.10.24.620086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Objective The capacity of the liver to serve as a peripheral sensor in the regulation of food intake has been debated for over half a century. The anatomical position and physiological roles of the liver suggest it is a prime candidate to serve as an interoceptive sensor of peripheral tissue and systemic energy state. Importantly, maintenance of liver ATP levels and within-meal food intake inhibition is impaired in human subjects with obesity and obese pre-clinical models. Previously, we have shown decreased hepatic mitochondrial energy metabolism (i.e., oxidative metabolism & ADP-dependent respiration) in male liver-specific, heterozygous PGC1a mice results in increased short-term diet-induced weight gain with increased within meal food intake. Herein, we tested the hypothesis that decreased liver mitochondrial energy metabolism impairs meal termination following nutrient oral pre-loads. Methods Liver mitochondrial respiratory response to changes in ΔGATP and adenine nucleotide concentration following fasting were examined in male liver-specific, heterozygous PGC1a mice. Further, food intake and feeding behavior during basal conditions, following nutrient oral pre-loads, and following fasting were investigated. Results We observed male liver-specific, heterozygous PGC1a mice have reduced mitochondrial response to changes in ΔGATP and tissue ATP following fasting. These impairments in liver energy state are associated with larger and longer meals during chow feeding, impaired dose-dependent food intake inhibition in response to mixed and individual nutrient oral pre-loads, and greater acute fasting-induced food intake. Conclusion These data support previous work proposing liver-mediated food intake regulation through modulation of peripheral satiation signals.
Collapse
Affiliation(s)
- Michael E. Ponte
- Dept. of Cell Biology & Physiology University of Kansas Medical Center, Kansas City, Kansas, USA
| | - John C. Prom
- Dept. of Cell Biology & Physiology University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Mallory A. Newcomb
- Dept. of Cell Biology & Physiology University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Annabelle B. Jordan
- Dept. of Cell Biology & Physiology University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Lucas L. Comfort
- Dept. of Cell Biology & Physiology University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Jiayin Hu
- Dept. of Psychiatry University of Pennsylvania, Philadelphia, PA, USA
| | - Patrycja Puchalska
- Division of Molecular Medicine, University of Minnesota, Minneapolis, MN
| | - Caroline E. Geisler
- Dept. of Psychiatry University of Pennsylvania, Philadelphia, PA, USA
- Dept. of Pharmaceutical Sciences University of Kentucky, Lexington, KY, USA
| | - Matthew R. Hayes
- Dept. of Psychiatry University of Pennsylvania, Philadelphia, PA, USA
| | - E. Matthew Morris
- Dept. of Cell Biology & Physiology University of Kansas Medical Center, Kansas City, Kansas, USA
- Center for Children’s Healthy Lifestyle and Nutrition Children’s Mercy Hospital, Kansas City, Missouri, USA
- University of Kansas Diabetes Institute Kansas City, Kansas
| |
Collapse
|
7
|
Fan YH, Zhang S, Wang Y, Wang H, Li H, Bai L. Inter-organ metabolic interaction networks in non-alcoholic fatty liver disease. Front Endocrinol (Lausanne) 2025; 15:1494560. [PMID: 39850476 PMCID: PMC11754069 DOI: 10.3389/fendo.2024.1494560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 12/16/2024] [Indexed: 01/25/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a multisystem metabolic disorder, marked by abnormal lipid accumulation and intricate inter-organ interactions, which contribute to systemic metabolic imbalances. NAFLD may progress through several stages, including simple steatosis (NAFL), non-alcoholic steatohepatitis (NASH), cirrhosis, and potentially liver cancer. This disease is closely associated with metabolic disorders driven by overnutrition, with key pathological processes including lipid dysregulation, impaired lipid autophagy, mitochondrial dysfunction, endoplasmic reticulum (ER) stress, and local inflammation. While hepatic lipid metabolism in NAFLD is well-documented, further research into inter-organ communication mechanisms is crucial for a deeper understanding of NAFLD progression. This review delves into intrahepatic networks and tissue-specific signaling mediators involved in NAFLD pathogenesis, emphasizing their impact on distal organs.
Collapse
Affiliation(s)
- Yu-Hong Fan
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Siyao Zhang
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Ye Wang
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Hongni Wang
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Hongliang Li
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Ganzhou, China
- Department of Cardiology, Renmin Hospital, Wuhan University, Wuhan, China
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lan Bai
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| |
Collapse
|
8
|
Mignini I, Galasso L, Piccirilli G, Calvez V, Termite F, Esposto G, Borriello R, Miele L, Ainora ME, Gasbarrini A, Zocco MA. Interplay of Oxidative Stress, Gut Microbiota, and Nicotine in Metabolic-Associated Steatotic Liver Disease (MASLD). Antioxidants (Basel) 2024; 13:1532. [PMID: 39765860 PMCID: PMC11727446 DOI: 10.3390/antiox13121532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 12/10/2024] [Accepted: 12/12/2024] [Indexed: 01/15/2025] Open
Abstract
Oxidative stress has been described as one of the main drivers of intracellular damage and metabolic disorders leading to metabolic syndrome, a major health problem worldwide. In particular, free radicals alter lipid metabolism and promote lipid accumulation in the liver, existing in the hepatic facet of metabolic syndrome, the metabolic dysfunction-associated steatotic liver disease (MASLD). Recent literature has highlighted how nicotine, especially if associated with a high-fat diet, exerts a negative effect on the induction and progression of MASLD by upregulating inflammation and increasing oxidative stress, abdominal fat lipolysis, and hepatic lipogenesis. Moreover, considerable evidence shows the central role of intestinal dysbiosis in the pathogenesis of MASLD and the impact of nicotine-induced oxidative stress on the gut microbiome. This results in an intricate network in which oxidative stress stands at the intersection point between gut microbiome, nicotine, and MASLD. The aim of this review is to delve into the molecular mechanisms linking tobacco smoking and MASLD, focusing on nicotine-induced microbiota modifications and their impact on MASLD development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Maria Assunta Zocco
- CEMAD Digestive Diseases Center, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy; (I.M.); (L.G.); (G.P.); (V.C.); (F.T.); (G.E.); (R.B.); (L.M.); (M.E.A.); (A.G.)
| |
Collapse
|
9
|
Lacal JC, Ibrahim SA, Zimmerman T. Is choline kinase alpha a drug target for obesity? Front Endocrinol (Lausanne) 2024; 15:1492753. [PMID: 39568820 PMCID: PMC11576158 DOI: 10.3389/fendo.2024.1492753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 10/15/2024] [Indexed: 11/22/2024] Open
Abstract
Choline kinase alpha (ChoKα) is a therapeutic target being developed for a variety of diseases, from cancer to rheumatoid arthritis and from parasites to bacterial infections. Nevertheless, the therapeutic potential of this drug target seems not exhausted and may end up as a possible solution for a larger variety of conditions. Here we present our working model for how ChoKα could play a role in obesity and for how drugs being developed as therapeutics for other diseases using ChoKα as a target, could be repurposed as prophylactic treatments for obesity. We also present preliminary observations in support of our model.
Collapse
Affiliation(s)
- Juan Carlos Lacal
- Department of Metabolic & Immune Diseases, Instituto de Investigaciones Biomédicas, Agencia Estatal Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Salam A. Ibrahim
- Food and Nutritional Sciences Program, Department of Family and Consumer Sciences, North Carolina Agricultural and Technical University, Greensboro, NC, United States
| | - Tahl Zimmerman
- Biomedical Sciences Program, High Point University, One University Parkway, High Point, NC, United States
| |
Collapse
|
10
|
Arconzo M, Piccinin E, Pasculli E, Cariello M, Loiseau N, Bertrand-Michel J, Guillou H, Matrella ML, Villani G, Moschetta A. Hepatic-specific Pgc-1α ablation drives fibrosis in a MASH model. Liver Int 2024; 44:2738-2752. [PMID: 39046166 DOI: 10.1111/liv.16052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 07/01/2024] [Accepted: 07/10/2024] [Indexed: 07/25/2024]
Abstract
BACKGROUND & AIMS Metabolic dysfunction-associated steatohepatitis (MASH) is a growing cause of chronic liver disease, characterized by fat accumulation, inflammation and fibrosis, which development depends on mitochondrial dysfunction and oxidative stress. Highly expressed in the liver during fasting, peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) regulates mitochondrial and oxidative metabolism. Given the relevant role of mitochondrial function in MASH, we investigated the relationship between PGC-1α and steatohepatitis. METHODS We measured the hepatic expression of Pgc-1α in both MASH patients and wild-type mice fed a western diet (WD) inducing steatosis and fibrosis. We then generated a pure C57BL6/J strain loss of function mouse model in which Pgc-1α is selectively deleted in the liver and we fed these mice with a WD supplemented with sugar water that accurately mimics human MASH. RESULTS We observed that the hepatic expression of Pgc-1α is strongly reduced in MASH, in both humans and mice. Moreover, the hepatic ablation of Pgc-1α promotes a considerable reduction of the hepatic mitochondrial respiratory capacity, setting up a bioenergetic harmful environment for liver diseases. Indeed, the lack of Pgc-1α decreases mitochondrial function and increases inflammation, fibrosis and oxidative stress in the scenario of MASH. Intriguingly, this profibrotic phenotype is not linked with obesity, insulin resistance and lipid disbalance. CONCLUSIONS In a MASH model the hepatic ablation of Pgc-1α drives fibrosis independently from lipid and glucose metabolism. These results add a novel mechanistic piece to the puzzle of the specific and crucial role of mitochondrial function in MASH development.
Collapse
Affiliation(s)
- Maria Arconzo
- Department of Interdisciplinary Medicine (DIM), University of Bari "Aldo Moro", Bari, Italy
| | - Elena Piccinin
- Department of Interdisciplinary Medicine (DIM), University of Bari "Aldo Moro", Bari, Italy
- Department of Translational Biomedicine and Neuroscience (DiBraiN), University of Bari "Aldo Moro", Bari, Italy
| | - Emanuela Pasculli
- Department of Interdisciplinary Medicine (DIM), University of Bari "Aldo Moro", Bari, Italy
| | - Marica Cariello
- Department of Interdisciplinary Medicine (DIM), University of Bari "Aldo Moro", Bari, Italy
| | - Nicolas Loiseau
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP-PURPAN, UMR 1331, UPS, Université de Toulouse, Toulouse, France
| | | | - Hervé Guillou
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP-PURPAN, UMR 1331, UPS, Université de Toulouse, Toulouse, France
| | - Maria L Matrella
- Department of Translational Biomedicine and Neuroscience (DiBraiN), University of Bari "Aldo Moro", Bari, Italy
| | - Gaetano Villani
- Department of Translational Biomedicine and Neuroscience (DiBraiN), University of Bari "Aldo Moro", Bari, Italy
| | - Antonio Moschetta
- Department of Interdisciplinary Medicine (DIM), University of Bari "Aldo Moro", Bari, Italy
- INBB, National Institute for Biostructures and Biosystems, Rome, Italy
| |
Collapse
|
11
|
Carvalho LCF, Ferreira FM, Dias BV, Azevedo DCD, de Souza GHB, Milagre MM, de Lana M, Vieira PMDA, Carneiro CM, Paula-Gomes SD, Cangussu SD, Costa DC. Silymarin inhibits the lipogenic pathway and reduces worsening of non-alcoholic fatty liver disease (NAFLD) in mice. Arch Physiol Biochem 2024; 130:460-474. [PMID: 36328030 DOI: 10.1080/13813455.2022.2138445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/17/2022] [Accepted: 09/08/2022] [Indexed: 11/06/2022]
Abstract
CONTEXT The role of silymarin in hepatic lipid dysfunction and its possible mechanisms of action were investigated. OBJECTIVE To evaluate the effects of silymarin on hepatic and metabolic profiles in mice fed with 30% fructose for 8 weeks. METHODS We evaluated the antioxidant profile of silymarin; mice consumed 30% fructose and were treated with silymarin (120 mg/kg/day or 240 mg/kg/day). We performed biochemical, redox status, and histopathological assays. RT-qPCR was performed to detect ACC-1, ACC-2, FAS, and CS expression, and western blotting to detect PGC-1α levels. RESULTS Silymarin contains high levels of phenolic compounds and flavonoids and exhibited significant antioxidant capacity in vitro. In vivo, the fructose-fed groups showed increased levels of AST, ALT, SOD/CAT, TBARS, hepatic TG, and cholesterol, as well as hypertriglyceridaemia, hypercholesterolaemia, and increased ACC-1 and FAS. Silymarin treatment reduced these parameters and increased mRNA levels and activity of hepatic citrate synthase. CONCLUSIONS These results suggest that silymarin reduces worsening of NAFLD.
Collapse
Affiliation(s)
| | | | - Bruna Vidal Dias
- Laboratório de Bioquímica Metabólica, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | | | | | - Matheus Marque Milagre
- Laboratório Doença de Chagas, Universidade Federal de Ouro Preto, UFOP, Ouro Preto, Brazil
| | - Marta de Lana
- Laboratório Doença de Chagas, Universidade Federal de Ouro Preto, UFOP, Ouro Preto, Brazil
| | | | | | - Sílvia de Paula-Gomes
- Laboratório de Bioquímica e Biologia Molecular, Universidade Federal de Ouro Preto, UFOP, Ouro Preto, Brazil
| | - Silvia Dantas Cangussu
- Laboratório de Fisiopatologia Experimental, Universidade Federal de Ouro Preto, UFOP, Ouro Preto, Brazil
| | - Daniela Caldeira Costa
- Laboratório de Bioquímica Metabólica, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| |
Collapse
|
12
|
Rajendran R, Suman S, Divakaran SJ, Swatikrishna S, Tripathi P, Jain R, Sagar K, Rajakumari S. Sesaminol alters phospholipid metabolism and alleviates obesity-induced NAFLD. FASEB J 2024; 38:e23835. [PMID: 39037555 DOI: 10.1096/fj.202400412rr] [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: 02/23/2024] [Revised: 07/04/2024] [Accepted: 07/10/2024] [Indexed: 07/23/2024]
Abstract
The prevalence of obesity-induced non-alcoholic fatty liver disease (NAFLD) and insulin resistance is increasing worldwide. We previously demonstrated that sesaminol increases thermogenesis in adipocytes, improves insulin sensitivity, and mitigates obesity in mice. In this study, we demonstrated that sesaminol increased mitochondrial activity and reduced ROS production in hepatocytes. Therefore, we delve into the metabolic action of sesaminol in obesity-induced NAFLD or metabolic dysfunction-associated liver disease (MAFLD). Here, we report that sesaminol induces OXPHOS proteins and mitochondrial function in vivo. Further, our data suggest that sesaminol administration reduces hepatic triacylglycerol accumulation and LDL-C levels. Prominently, the lipidomics analyses revealed that sesaminol administration decreased the major phospholipids such as PC, PE, PI, CL, and PS to maintain membrane lipid homeostasis in the liver upon HFD challenge. Besides, SML reduced ePC and SM molecular species and increased PA levels in the HFD-fed mice. Also, sesaminol renders anti-inflammatory properties and dampens fibrosis markers in the liver. Remarkably, SML lowers the hepatic levels of ALT and AST enzymes and alleviates NAFLD in diet-induced obese mice. The molecular docking analysis identifies peroxisome proliferator-activated receptors as potential endogenous receptors for sesaminol. Together, our study demonstrates plant lignan sesaminol as a potential small molecule that alters the molecular species of major phospholipids, including sphingomyelin and ether-linked PCs in the liver tissue, improves metabolic parameters, and alleviates obesity-induced fatty liver disease in mice.
Collapse
Affiliation(s)
- Rajprabu Rajendran
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru, Karnataka, India
| | - Sanskriti Suman
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru, Karnataka, India
| | - Soumya Jaya Divakaran
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru, Karnataka, India
| | - Sahu Swatikrishna
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru, Karnataka, India
| | - Purnima Tripathi
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru, Karnataka, India
| | - Rashi Jain
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru, Karnataka, India
| | - Karan Sagar
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru, Karnataka, India
| | - Sona Rajakumari
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru, Karnataka, India
| |
Collapse
|
13
|
Caon E, Martins M, Hodgetts H, Blanken L, Vilia MG, Levi A, Thanapirom K, Al-Akkad W, Abu-Hanna J, Baselli G, Hall AR, Luong TV, Taanman JW, Vacca M, Valenti L, Romeo S, Mazza G, Pinzani M, Rombouts K. Exploring the impact of the PNPLA3 I148M variant on primary human hepatic stellate cells using 3D extracellular matrix models. J Hepatol 2024; 80:941-956. [PMID: 38365182 DOI: 10.1016/j.jhep.2024.01.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/24/2024] [Accepted: 01/27/2024] [Indexed: 02/18/2024]
Abstract
BACKGROUND & AIMS The PNPLA3 rs738409 C>G (encoding for I148M) variant is a risk locus for the fibrogenic progression of chronic liver diseases, a process driven by hepatic stellate cells (HSCs). We investigated how the PNPLA3 I148M variant affects HSC biology using transcriptomic data and validated findings in 3D-culture models. METHODS RNA sequencing was performed on 2D-cultured primary human HSCs and liver biopsies of individuals with obesity, genotyped for the PNPLA3 I148M variant. Data were validated in wild-type (WT) or PNPLA3 I148M variant-carrying HSCs cultured on 3D extracellular matrix (ECM) scaffolds from human healthy and cirrhotic livers, with/without TGFB1 or cytosporone B (Csn-B) treatment. RESULTS Transcriptomic analyses of liver biopsies and HSCs highlighted shared PNPLA3 I148M-driven dysregulated pathways related to mitochondrial function, antioxidant response, ECM remodelling and TGFB1 signalling. Analogous pathways were dysregulated in WT/PNPLA3-I148M HSCs cultured in 3D liver scaffolds. Mitochondrial dysfunction in PNPLA3-I148M cells was linked to respiratory chain complex IV insufficiency. Antioxidant capacity was lower in PNPLA3-I148M HSCs, while reactive oxygen species secretion was increased in PNPLA3-I148M HSCs and higher in bioengineered cirrhotic vs. healthy scaffolds. TGFB1 signalling followed the same trend. In PNPLA3-I148M cells, expression and activation of the endogenous TGFB1 inhibitor NR4A1 were decreased: treatment with the Csn-B agonist increased total NR4A1 in HSCs cultured in healthy but not in cirrhotic 3D scaffolds. NR4A1 regulation by TGFB1/Csn-B was linked to Akt signalling in PNPLA3-WT HSCs and to Erk signalling in PNPLA3-I148M HSCs. CONCLUSION HSCs carrying the PNPLA3 I148M variant have impaired mitochondrial function, antioxidant responses, and increased TGFB1 signalling, which dampens antifibrotic NR4A1 activity. These features are exacerbated by cirrhotic ECM, highlighting the dual impact of the PNPLA3 I148M variant and the fibrotic microenvironment in progressive chronic liver diseases. IMPACT AND IMPLICATIONS Hepatic stellate cells (HSCs) play a key role in the fibrogenic process associated with chronic liver disease. The PNPLA3 genetic mutation has been linked with increased risk of fibrogenesis, but its role in HSCs requires further investigation. Here, by using comparative transcriptomics and a novel 3D in vitro model, we demonstrate the impact of the PNPLA3 genetic mutation on primary human HSCs' behaviour, and we show that it affects the cell's mitochondrial function and antioxidant response, as well as the antifibrotic gene NR4A1. Our publicly available transcriptomic data, 3D platform and our findings on NR4A1 could facilitate the discovery of targets to develop more effective treatments for chronic liver diseases.
Collapse
Affiliation(s)
- Elisabetta Caon
- Regenerative Medicine and Fibrosis Group, Institute for Liver and Digestive Health, University College London, Royal Free Campus, London, UK
| | - Maria Martins
- Regenerative Medicine and Fibrosis Group, Institute for Liver and Digestive Health, University College London, Royal Free Campus, London, UK
| | - Harry Hodgetts
- Regenerative Medicine and Fibrosis Group, Institute for Liver and Digestive Health, University College London, Royal Free Campus, London, UK
| | - Lieke Blanken
- Regenerative Medicine and Fibrosis Group, Institute for Liver and Digestive Health, University College London, Royal Free Campus, London, UK
| | - Maria Giovanna Vilia
- Regenerative Medicine and Fibrosis Group, Institute for Liver and Digestive Health, University College London, Royal Free Campus, London, UK
| | - Ana Levi
- Regenerative Medicine and Fibrosis Group, Institute for Liver and Digestive Health, University College London, Royal Free Campus, London, UK
| | - Kessarin Thanapirom
- Regenerative Medicine and Fibrosis Group, Institute for Liver and Digestive Health, University College London, Royal Free Campus, London, UK
| | - Walid Al-Akkad
- Regenerative Medicine and Fibrosis Group, Institute for Liver and Digestive Health, University College London, Royal Free Campus, London, UK
| | - Jeries Abu-Hanna
- Research Department of Surgical Biotechnology, Division of Surgery and Interventional Science, University College London, London, UK
| | - Guido Baselli
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Andrew R Hall
- Sheila Sherlock Liver Centre, Royal Free London NHS Foundation Trust, London, UK; Department of Cellular Pathology, Royal Free London NHS Foundation Trust, London, UK
| | - Tu Vinh Luong
- Sheila Sherlock Liver Centre, Royal Free London NHS Foundation Trust, London, UK; Department of Cellular Pathology, Royal Free London NHS Foundation Trust, London, UK
| | - Jan-Willem Taanman
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London UK
| | - Michele Vacca
- Laboratory of Hepatic Metabolism and NAFLD, Roger Williams Institute of Hepatology, London, UK; Clinica Medica "Frugoni", Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy; Precision Medicine, Biological Resource Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefano Romeo
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| | - Giuseppe Mazza
- Regenerative Medicine and Fibrosis Group, Institute for Liver and Digestive Health, University College London, Royal Free Campus, London, UK
| | - Massimo Pinzani
- Regenerative Medicine and Fibrosis Group, Institute for Liver and Digestive Health, University College London, Royal Free Campus, London, UK
| | - Krista Rombouts
- Regenerative Medicine and Fibrosis Group, Institute for Liver and Digestive Health, University College London, Royal Free Campus, London, UK.
| |
Collapse
|
14
|
Aguilar EC, Fernandes-Braga W, Santos EA, Leocádio PCL, Dos Santos Aggum Capettini L, Orellano LAA, Campos PP, Lemos VS, Soares FLP, Navia-Pelaez JM, Alvarez-Leite JI. Gluten worsens non-alcoholic fatty liver disease by affecting lipogenesis and fatty acid oxidation in diet-induced obese apolipoprotein E-deficient mice. Mol Cell Biochem 2024; 479:1335-1347. [PMID: 37402020 DOI: 10.1007/s11010-023-04802-3] [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: 04/23/2023] [Accepted: 06/25/2023] [Indexed: 07/05/2023]
Abstract
Obesity is closely associated with non-alcoholic fatty liver disease (NAFLD), characterized by hepatic fat accumulation and hepatocyte injury. Preclinical studies have shown exacerbated weight gain associated with an obesogenic gluten-containing diet. However, whether gluten affects obesity-induced hepatic lipid accumulation still remains unclear. We hypothesized that gluten intake could affect fatty liver development in high-fat diet (HFD)-induced obese mice. Thus, we aimed to investigate the impact of gluten intake on NAFLD in HFD-induced obese mice. Male apolipoprotein E-deficient (Apoe-/-) mice were fed with a HFD containing (GD) or not (GFD) vital wheat gluten (4.5%) for 10 weeks. Blood and liver were collected for further analysis. We found that gluten exacerbated weight gain, hepatic fat deposition, and hyperglycemia without affecting the serum lipid profile. Livers of the GD group showed a larger area of fibrosis, associated with the expression of collagen and MMP9, and higher expression of apoptosis-related factors, p53, p21, and caspase-3. The expression of lipogenic factors, such as PPARγ and Acc1, was more elevated and factors related to beta-oxidation, such as PPARα and Cpt1, were lower in the GD group compared to the GFD. Further, gluten intake induced a more significant expression of Cd36, suggesting higher uptake of free fatty acids. Finally, we found lower protein expression of PGC1α followed by lower activation of AMPK. Our data show that gluten-containing high-fat diet exacerbated NAFLD by affecting lipogenesis and fatty acid oxidation in obese Apoe-/- mice through a mechanism involving lower activation of AMPK.
Collapse
Affiliation(s)
- Edenil Costa Aguilar
- Department of Biochemistry and Immunology, ICB - Federal University of Minas Gerais, Caixa Postal 486, Belo Horizonte, 30161-970, Brazil.
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Weslley Fernandes-Braga
- Department of Biochemistry and Immunology, ICB - Federal University of Minas Gerais, Caixa Postal 486, Belo Horizonte, 30161-970, Brazil
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Elandia Aparecida Santos
- Department of Biochemistry and Immunology, ICB - Federal University of Minas Gerais, Caixa Postal 486, Belo Horizonte, 30161-970, Brazil
| | - Paola Caroline Lacerda Leocádio
- Department of Biochemistry and Immunology, ICB - Federal University of Minas Gerais, Caixa Postal 486, Belo Horizonte, 30161-970, Brazil
| | | | | | - Paula Peixoto Campos
- Department of General Pathology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Virginia Soares Lemos
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Juliana Maria Navia-Pelaez
- Department of Pharmacology, Federal University of Minas Gerais, Belo Horizonte, Brazil
- Department of Medicine, University of California San Diego, San Diego, USA
| | - Jacqueline I Alvarez-Leite
- Department of Biochemistry and Immunology, ICB - Federal University of Minas Gerais, Caixa Postal 486, Belo Horizonte, 30161-970, Brazil
| |
Collapse
|
15
|
Yuan XM, Xiang MQ, Ping Y, Zhang PW, Liu YT, Liu XW, Wei J, Tang Q, Zhang Y. Beneficial Effects of High-Intensity Interval Training and Dietary Changes Intervention on Hepatic Fat Accumulation in HFD-Induced Obese Rats. Physiol Res 2024; 73:273-284. [PMID: 38710057 PMCID: PMC11081183 DOI: 10.33549/physiolres.935195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 11/02/2023] [Indexed: 01/05/2025] Open
Abstract
Lifestyle intervention encompassing nutrition and physical activity are effective strategies to prevent progressive lipid deposition in the liver. This study aimed to explore the effect of dietary change, and/or high-intensity interval training (HIIT) on hepatic lipid accumulation in high fat diet (HFD)-induced obese rats. We divided lean rats into lean control (LC) or HIIT groups (LH), and obese rats into obese normal chow diet (ND) control (ONC) or HIIT groups (ONH) and obese HFD control (OHC) or HIIT groups (OHH). We found that dietary or HIIT intervention significantly decreased body weight and the risk of dyslipidemia, prevented hepatic lipid accumulation. HIIT significantly improved mitochondrial fatty acid oxidation through upregulating mitochondrial enzyme activities, mitochondrial function and AMPK/PPARalpha/CPT1alpha pathway, as well as inhibiting hepatic de novo lipogenesis in obese HFD rats. These findings indicate that dietary alone or HIIT intervention powerfully improve intrahepatic storage of fat in diet induced obese rats. Keywords: Obesity, Exercise, Diet, Mitochondrial function, Lipid deposition.
Collapse
Affiliation(s)
- X-M Yuan
- School of Sports and Health, Nanjing Sport Institute, Nanjing, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Sun J, Chen Y, Wang T, Ali W, Ma Y, Yuan Y, Gu J, Bian J, Liu Z, Zou H. Baicalin and N-acetylcysteine regulate choline metabolism via TFAM to attenuate cadmium-induced liver fibrosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 125:155337. [PMID: 38241915 DOI: 10.1016/j.phymed.2024.155337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/14/2023] [Accepted: 01/05/2024] [Indexed: 01/21/2024]
Abstract
(Background): Cadmium is an environmental pollutant associated with several liver diseases. Baicalin and N-Acetylcysteine have antioxidant and hepatoprotective effects. (Purpose): However, it is unclear whether baicalin and N-Acetylcysteine can alleviate Cadmium -induced liver fibrosis by regulating metabolism, or whether they exert a synergistic effect. (Study design): We treated Cadmium-poisoned mice with baicalin, N-Acetylcysteine, or baicalin+ N-Acetylcysteine. We studied the effects of baicalin and N-Acetylcysteine on Cadmium-induced liver fibers and their specific mechanisms. (Methods): We used C57BL/6 J mice, and AML12, and HSC-6T cells to establish in vitro assays and in vivo models. (Results): Metabolomics was used to detect the effect of baicalin and N-Acetylcysteine on liver metabolism, which showed that compared with the control group, the Cadmium group had increased fatty acid and amino acid levels, with significantly reduced choline and acetylcholine contents. Baicalin and N-Acetylcysteine alleviated these Cadmium-induced metabolic changes. We further showed that choline alleviated Cadmium -induced liver inflammation and fibrosis. In addition, cadmium significantly promoted extracellular leakage of lactic acid, while choline alleviated the cadmium -induced destruction of the cell membrane structure and lactic acid leakage. Western blotting showed that cadmium significantly reduced mitochondrial transcription factor A (TFAM) and Choline Kinase α(CHKα2) levels, and baicalin and N-Acetylcysteine reversed this effect. Overexpression of Tfam in mouse liver and AML12 cells increased the expression of CHKα2 and the choline content, alleviating and cadmium-induced lactic acid leakage, liver inflammation, and fibrosis. (Conclusion): Overall, baicalin and N-Acetylcysteine alleviated cadmium-induced liver damage, inflammation, and fibrosis to a greater extent than either drug alone. TFAM represents a target for baicalin and N-Acetylcysteine, and alleviated cadmium-induced liver inflammation and fibrosis by regulating hepatic choline metabolism.
Collapse
Affiliation(s)
- Jian Sun
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China
| | - Yan Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China
| | - Tao Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China
| | - Waseem Ali
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China
| | - Yonggang Ma
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yan Yuan
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Jianhong Gu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Jianchun Bian
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Zongping Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Hui Zou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.
| |
Collapse
|
17
|
Hu J, Chen Z, Zhou Y, Li Y, Liu J, Mi Y, Wang L, Jiang F, Li P. Unveiling global research trends and hotspots on mitochondria in NAFLD from 2000 to 2023: A bibliometric analysis. Immun Inflamm Dis 2024; 12:e1226. [PMID: 38533910 PMCID: PMC10966917 DOI: 10.1002/iid3.1226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/01/2024] [Accepted: 03/08/2024] [Indexed: 03/28/2024] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) has garnered significant attention in the past decade as a prevalent chronic liver condition. Despite a growing body of evidence implicating mitochondria in NAFLD development, comprehensive bibliometric analyses within this research domain are scarce. This study aims to provide a thorough overview of the knowledge framework and key research areas related to mitochondria in the context of NAFLD, utilizing bibliometric techniques. METHODS A comprehensive search of publications on mitochondria in NAFLD from 2000 to 2023 was conducted using the Web of Science Core Collection database. VOSviewers, CiteSpace, and the R package "bibliometrix" were employed for a precise assessment of the literature. RESULTS Examining 2530 articles from 77 countries, primarily led by the United States and China, revealed a consistent increase in publications on mitochondria's role in NAFLD. Leading research institutions include the University of Coimbra, the University of Missouri, the Chinese Academy of Sciences, Fudan University, and Shanghai Jiao Tong University. Notably, the International Journal of Molecular Sciences emerged as the most popular journal, and Hepatology was the most frequently cited. With contributions from 14,543 authors, Michael Roden published the highest number of papers, and A. J. Samyal was the most frequently cocited author. Key focus areas include investigating mitochondrial mechanisms impacting NAFLD and developing therapeutic strategies targeting mitochondria. Emerging research hotspots are associated with keywords such as "inflammation," "mitochondrial dysfunction," "autophagy," "obesity," and "insulin resistance." CONCLUSION This study, the first comprehensive bibliometric analysis, synthesizes research trends and advancements in the role of mitochondria in NAFLD. Insights derived from this analysis illuminate current frontiers and emerging areas of interest, providing a valuable reference for scholars dedicated to mitochondrial studies.
Collapse
Affiliation(s)
- Jingqin Hu
- Clinical School of the Second People's HospitalTianjin Medical UniversityTianjinChina
- Department of HepatologyTianjin Second People's HospitalTianjinChina
| | - Ze Chen
- Clinical School of the Second People's HospitalTianjin Medical UniversityTianjinChina
- Department of HepatologyTianjin Second People's HospitalTianjinChina
| | - Yibing Zhou
- Clinical School of the Second People's HospitalTianjin Medical UniversityTianjinChina
- Department of HepatologyTianjin Second People's HospitalTianjinChina
| | - Yinglun Li
- Clinical School of the Second People's HospitalTianjin Medical UniversityTianjinChina
- Department of HepatologyTianjin Second People's HospitalTianjinChina
| | - Jing Liu
- Clinical School of the Second People's HospitalTianjin Medical UniversityTianjinChina
- Department of HepatologyTianjin Second People's HospitalTianjinChina
| | - Yuqiang Mi
- Department of HepatologyTianjin Second People's HospitalTianjinChina
| | - Li Wang
- Department of PharmacyTianjin Second People's HospitalTianjinChina
| | - Feng Jiang
- Department of NeonatologyObstetrics and Gynecology Hospital of Fudan UniversityShanghaiChina
| | - Ping Li
- Department of HepatologyTianjin Second People's HospitalTianjinChina
| |
Collapse
|
18
|
Placha W, Suder P, Panek A, Bronowicka-Adamska P, Zarzycka M, Szczygieł M, Zagajewski J, Piwowar MW. The Blocking of Drug Resistance Channels by Selected Hydrophobic Statins in Chemoresistance Human Melanoma. Biomolecules 2023; 13:1682. [PMID: 38136555 PMCID: PMC10741734 DOI: 10.3390/biom13121682] [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: 10/06/2023] [Revised: 11/09/2023] [Accepted: 11/17/2023] [Indexed: 12/24/2023] Open
Abstract
Despite the development of modern drugs, drug resistance in oncology remains the main factor limiting the curability of patients. This paper shows the use of a group of hydrophobic statins to inhibit drug resistance (Pgp protein). In a chemoresistance melanoma cell model, viability, necroptosis with DNA damage, the absorption of the applied pharmaceuticals, and the functional activity of the ABCB1 drug transporter after administration of docetaxel or docetaxel with a selected hydrophobic statin were studied. Taxol-resistant human melanoma cells from three stages of development were used as a model: both A375P and WM239A metastatic lines and radial growth phase WM35 cells. An animal model (Mus musculus SCID) was developed for the A375P cell line. The results show that hydrophobic statins administered with docetaxel increase the accumulation of the drug in the tumor cell a.o. by blocking the ABCB1 channel. They reduce taxol-induced drug resistance. The tumor size reduction was observed after the drug combination was administrated. It was shown that the structural similarity of statins is of secondary importance, e.g., pravastatin and simvastatin. Using cytostatics in the presence of hydrophobic statins increases their effectiveness while reducing their overall toxicity.
Collapse
Affiliation(s)
- Wojciech Placha
- Department of Biochemistry, Faculty of Medicine, Jagiellonian University Medical College, Kopernika 7b St., 31-034 Krakow, Poland; (P.B.-A.); (M.Z.); (J.Z.)
| | - Piotr Suder
- Department of Analytical Chemistry and Biochemistry, Faculty of Materials Science and Ceramics, AGH University of Science and Technology, 31-007 Krakow, Poland;
| | - Agnieszka Panek
- Institute of Nuclear Physics Polish Academy of Sciences, 31-342 Krakow, Poland;
| | - Patrycja Bronowicka-Adamska
- Department of Biochemistry, Faculty of Medicine, Jagiellonian University Medical College, Kopernika 7b St., 31-034 Krakow, Poland; (P.B.-A.); (M.Z.); (J.Z.)
| | - Marta Zarzycka
- Department of Biochemistry, Faculty of Medicine, Jagiellonian University Medical College, Kopernika 7b St., 31-034 Krakow, Poland; (P.B.-A.); (M.Z.); (J.Z.)
| | - Małgorzata Szczygieł
- Department of Biophysics and Cancer Biology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, 31-007 Krakow, Poland;
| | - Jacek Zagajewski
- Department of Biochemistry, Faculty of Medicine, Jagiellonian University Medical College, Kopernika 7b St., 31-034 Krakow, Poland; (P.B.-A.); (M.Z.); (J.Z.)
| | - Monika Weronika Piwowar
- Department of Bioinformatics and Telemedicine, Faculty of Medicine, Jagiellonian University Medical College, Kopernika 7e St., 31-034 Krakow, Poland;
| |
Collapse
|
19
|
Song J, Ren L, Ren Z, Ren X, Qi Y, Qin Y, Zhang X, Ren Y, Li Y. SIRT1-dependent mitochondrial biogenesis supports therapeutic effects of 4-butyl-polyhydroxybenzophenone compounds against NAFLD. Eur J Med Chem 2023; 260:115728. [PMID: 37625288 DOI: 10.1016/j.ejmech.2023.115728] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 08/12/2023] [Accepted: 08/13/2023] [Indexed: 08/27/2023]
Abstract
The mitochondria have been identified as key targets in nonalcoholic fatty liver disease (NAFLD), one of the most prevalent chronic liver damage diseases globally. Meanwhile, the biological information analysis in this study revealed that SIRT1, PPARG, PPARA, and PPARGC1A (mitochondrial biogenesis-related proteins) were NAFLD therapeutic targets. Therefore, the design and synthesis of targeted drugs that promote mitochondrial biogenesis and improve mitochondrial function are particularly important for NAFLD treatment. Recently, we introduced butyls, hydroxyls, and halogens to benzophenone and synthesized a series of NAFLD-related 4-butylpolyhydroxybenzophenone compounds, aiming at investigating the hepatoprotective activity from the aspect of mitochondrial biogenesis. The structure-activity relationship demonstrated that hydroxyl and ketone groups were active groups interacting with mitochondrial biogenesis proteins (SIRT1 and PGC1α), and the activity was stronger when the o-hydroxyl group was present on the benzene ring. In contrast, the activity was little affected by the presence of the p-hydroxyl group, m-hydroxyl group, butyl group type, or halogen. In addition, in vitro studies confirmed that these compounds could directly bind to SIRT1 and PGC1α, markedly promote their interaction, significantly increase the expression of proteins and genes related to mitochondrial biogenesis (SIRT1, PGC1α, NRF1, TFAM, COX1, and ND6) and subsequently ameliorate mitochondria dysfunction, which was evidenced by the decreased ROS, upregulated ATP production, increased MMP, and enhanced mitochondrial number. According to the outcomes of our in vitro and in vivo experiments, 4-butyl-polyhydroxybenzophenone compounds could also effectively reduce the formation of lipid droplets and liver injury index (ALT, AST, LDH, AKP, γ-GT, and GDH) and improve the level of antioxidant enzymes (GSH and SOD). Particularly, the treatment of these compounds after a high-fat diet could significantly reduce body weight, decrease liver coefficient, attenuate liver damage, and ameliorate lipid accumulation in rat liver, demonstrating their therapeutic effects on NAFLD. Mechanistically, 4-butyl-polyhydroxybenzophenone compounds promoted mitochondrial biogenesis and eventually prevented NAFLD liver injury by activating the PGC1α signaling pathway in a SIRT1-dependent manner, which was strongly supported by SIRT1 inhibitor EX527.
Collapse
Affiliation(s)
- Jiayu Song
- School of Pharmaceutical Science, Shanxi Medical University, Taiyuan, 030001, PR China
| | - Luyao Ren
- School of Pharmaceutical Science, Shanxi Medical University, Taiyuan, 030001, PR China
| | - Zhenzhu Ren
- School of Pharmaceutical Science, Shanxi Medical University, Taiyuan, 030001, PR China
| | - Xing Ren
- School of Pharmaceutical Science, Shanxi Medical University, Taiyuan, 030001, PR China
| | - Yang Qi
- School of Pharmaceutical Science, Shanxi Medical University, Taiyuan, 030001, PR China
| | - Yuxi Qin
- School of Pharmaceutical Science, Shanxi Medical University, Taiyuan, 030001, PR China
| | - Xiaohui Zhang
- School of Pharmaceutical Science, Shanxi Medical University, Taiyuan, 030001, PR China
| | - Yuan Ren
- School of Pharmaceutical Science, Shanxi Medical University, Taiyuan, 030001, PR China
| | - Yunlan Li
- School of Pharmaceutical Science, Shanxi Medical University, Taiyuan, 030001, PR China; School of Public Health, Shaanxi University of Chinese Medicine, Xi'an, 712046, PR China.
| |
Collapse
|
20
|
Abrahams Y, Willmer T, Patel O, Samodien E, Muller CJF, Windvogel S, Johnson R, Pheiffer C. A high fat, high sugar diet induces hepatic Peroxisome proliferator-activated receptor gamma coactivator 1-alpha promoter hypermethylation in male Wistar rats. Biochem Biophys Res Commun 2023; 680:25-33. [PMID: 37713959 DOI: 10.1016/j.bbrc.2023.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/31/2023] [Accepted: 09/02/2023] [Indexed: 09/17/2023]
Abstract
Previously we reported that a high fat, high sugar (HFHS) diet induced adiposity, hyperinsulinaemia, hyperleptinaemia, hypertriglyceridaemia and increased liver mass in male Wistar rats. In the present study, the mechanisms underlying the increased liver mass were further elucidated by assessing hepatic lipid accumulation and the expression and methylation status of key metabolic genes using histology, quantitative real-time PCR and pyrosequencing, respectively. The HFHS diet induced hepatic steatosis, increased hepatic triglycerides (1.8-fold, p < 0.001), and increased the expression of sterol regulatory element-binding transcription factor 1 (Srebf1) (2.0-fold, p < 0.001) and peroxisome proliferator-activated receptor gamma (Pparg) (1.7-fold, p = 0.017) in the liver. The expression of peroxisome proliferator-activated receptor gamma coactivator 1 alpha (Pgc1a) was decreased (2.6-fold, p < 0.010), which was accompanied by hypermethylation (p = 0.018) of a conserved CpG site in the promoter of Pgc1a in HFHS fed rats compared to controls. In silico analysis identified putative binding sites for CCAAT/enhancer-binding protein beta (C/EBPß) and hepatocyte nuclear factor 1 (HNF1) within proximity to the hypermethylated CpG. As Pgc1a is a co-activator of several transcription factors regulating multiple metabolic pathways, hypermethylation of this conserved CpG site in the promoter of Pgc1a may be one possible mechanism contributing to the development of hepatic steatosis in response to a HFHS diet. However, further work is required to confirm the role of Pgc1a in steatosis.
Collapse
Affiliation(s)
- Yoonus Abrahams
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa; Centre for Cardio-Metabolic Research in Africa (CARMA), Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Tarryn Willmer
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa; Centre for Cardio-Metabolic Research in Africa (CARMA), Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa; Division of Cell Biology, Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Oelfah Patel
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa; Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, University of Stellenbosch, Tygerberg, South Africa
| | - Ebrahim Samodien
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
| | - Christo J F Muller
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa; Centre for Cardio-Metabolic Research in Africa (CARMA), Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa; Department of Biochemistry and Microbiology, University of Zululand, Kwadlangezwa, South Africa
| | - Shantal Windvogel
- Centre for Cardio-Metabolic Research in Africa (CARMA), Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Rabia Johnson
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa; Centre for Cardio-Metabolic Research in Africa (CARMA), Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Carmen Pheiffer
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa; Centre for Cardio-Metabolic Research in Africa (CARMA), Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa; Department of Obstetrics and Gynaecology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa.
| |
Collapse
|
21
|
Zhang T, Nie Y, Wang J. The emerging significance of mitochondrial targeted strategies in NAFLD treatment. Life Sci 2023; 329:121943. [PMID: 37454757 DOI: 10.1016/j.lfs.2023.121943] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/04/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most prevalent chronic liver disease worldwide, ranging from liver steatosis to nonalcoholic steatohepatitis, which ultimately progresses to fibrosis, cirrhosis, and hepatocellular carcinoma. Individuals with NAFLD have a higher risk of developing cardiovascular and extrahepatic cancers. Despite the great progress being made in understanding the pathogenesis and the introduction of new pharmacological targets for NAFLD, no drug or intervention has been accepted for its management. Recent evidence suggests that NAFLD may be a mitochondrial disease, as mitochondrial dysfunction is involved in the pathological processes that lead to NAFLD. In this review, we describe the recent advances in our understanding of the mechanisms associated with mitochondrial dysfunction in NAFLD progression. Moreover, we discuss recent advances in the efficacy of mitochondria-targeted compounds (e.g., Mito-Q, MitoVit-E, MitoTEMPO, SS-31, mitochondrial uncouplers, and mitochondrial pyruvate carrier inhibitors) for treating NAFLD. Furthermore, we present some medications currently being tested in clinical trials for NAFLD treatment, such as exercise, mesenchymal stem cells, bile acids and their analogs, and antidiabetic drugs, with a focus on their efficacy in improving mitochondrial function. Based on this evidence, further investigations into the development of mitochondria-based agents may provide new and promising alternatives for NAFLD management.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Yingli Nie
- Department of Dermatology, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China.
| | - Jiliang Wang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
22
|
Robea MA, Balmus IM, Girleanu I, Huiban L, Muzica C, Ciobica A, Stanciu C, Cimpoesu CD, Trifan A. Coagulation Dysfunctions in Non-Alcoholic Fatty Liver Disease-Oxidative Stress and Inflammation Relevance. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1614. [PMID: 37763733 PMCID: PMC10535217 DOI: 10.3390/medicina59091614] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/29/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the most common liver diseases. Its incidence is progressively rising and it is possibly becoming a worldwide epidemic. NAFLD encompasses a spectrum of diseases accounting for the chronic accumulation of fat within the hepatocytes due to various causes, excluding excessive alcohol consumption. In this study, we aimed to focus on finding evidence regarding the implications of oxidative stress and inflammatory processes that form the multifaceted pathophysiological tableau in relation to thrombotic events that co-occur in NAFLD and associated chronic liver diseases. Recent evidence on the pathophysiology of NAFLD suggests that a complex pattern of multidirectional components, such as prooxidative, proinflammatory, and prothrombotic components, better explains the multiple factors that promote the mechanisms underlying the fatty acid excess and subsequent processes. As there is extensive evidence on the multi-component nature of NAFLD pathophysiology, further studies could address the complex interactions that underlie the development and progression of the disease. Therefore, this study aimed to describe possible pathophysiological mechanisms connecting the molecular impairments with the various clinical manifestations, focusing especially on the interactions among oxidative stress, inflammation, and coagulation dysfunctions. Thus, we described the possible bidirectional modulation among coagulation homeostasis, oxidative stress, and inflammation that occurs in the various stages of NAFLD.
Collapse
Affiliation(s)
- Madalina Andreea Robea
- CENEMED Platform for Interdisciplinary Research, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.A.R.); (I.-M.B.); (C.D.C.)
| | - Ioana-Miruna Balmus
- CENEMED Platform for Interdisciplinary Research, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.A.R.); (I.-M.B.); (C.D.C.)
- Department of Exact Sciences and Natural Sciences, Institute of Interdisciplinary Research, “Alexandru Ioan Cuza” University of Iasi, Alexandru Lapusneanu Street, No. 26, 700057 Iasi, Romania
| | - Irina Girleanu
- Department of Gastroenterology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (I.G.); (L.H.); (C.M.); (A.T.)
- Institute of Gastroenterology and Hepatology, “St. Spiridon” University Hospital, 700111 Iasi, Romania
| | - Laura Huiban
- Department of Gastroenterology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (I.G.); (L.H.); (C.M.); (A.T.)
- Institute of Gastroenterology and Hepatology, “St. Spiridon” University Hospital, 700111 Iasi, Romania
| | - Cristina Muzica
- Department of Gastroenterology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (I.G.); (L.H.); (C.M.); (A.T.)
- Institute of Gastroenterology and Hepatology, “St. Spiridon” University Hospital, 700111 Iasi, Romania
| | - Alin Ciobica
- Department of Biology, Faculty of Biology, “Alexandru Ioan Cuza” University, Carol I Avenue, No. 20A, 700505 Iasi, Romania
- Centre of Biomedical Research, Romanian Academy, Carol I Avenue, No. 8, 700506 Iasi, Romania;
- Academy of Romanian Scientists, Splaiul Independentei nr. 54, Sector 5, 050094 Bucuresti, Romania
| | - Carol Stanciu
- Centre of Biomedical Research, Romanian Academy, Carol I Avenue, No. 8, 700506 Iasi, Romania;
| | - Carmen Diana Cimpoesu
- CENEMED Platform for Interdisciplinary Research, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.A.R.); (I.-M.B.); (C.D.C.)
- Department of Emergency Medicine, Emergency County Hospital “Sf. Spiridon”, 700111 Iasi, Romania
- Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa” Iasi, Blvd. Independentei 1, 700111 Iasi, Romania
| | - Anca Trifan
- Department of Gastroenterology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (I.G.); (L.H.); (C.M.); (A.T.)
- Institute of Gastroenterology and Hepatology, “St. Spiridon” University Hospital, 700111 Iasi, Romania
- Centre of Biomedical Research, Romanian Academy, Carol I Avenue, No. 8, 700506 Iasi, Romania;
| |
Collapse
|
23
|
Amorim R, Magalhães CC, Borges F, Oliveira PJ, Teixeira J. From Non-Alcoholic Fatty Liver to Hepatocellular Carcinoma: A Story of (Mal)Adapted Mitochondria. BIOLOGY 2023; 12:biology12040595. [PMID: 37106795 PMCID: PMC10135755 DOI: 10.3390/biology12040595] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 03/30/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a global pandemic affecting 25% of the world's population and is a serious health and economic concern worldwide. NAFLD is mainly the result of unhealthy dietary habits combined with sedentary lifestyle, although some genetic contributions to NAFLD have been documented. NAFLD is characterized by the excessive accumulation of triglycerides (TGs) in hepatocytes and encompasses a spectrum of chronic liver abnormalities, ranging from simple steatosis (NAFL) to steatohepatitis (NASH), significant liver fibrosis, cirrhosis, and hepatocellular carcinoma. Although the molecular mechanisms that cause the progression of steatosis to severe liver damage are not fully understood, metabolic-dysfunction-associated fatty liver disease is strong evidence that mitochondrial dysfunction plays a significant role in the development and progression of NAFLD. Mitochondria are highly dynamic organelles that undergo functional and structural adaptations to meet the metabolic requirements of the cell. Alterations in nutrient availability or cellular energy needs can modify mitochondria formation through biogenesis or the opposite processes of fission and fusion and fragmentation. In NAFL, simple steatosis can be seen as an adaptive response to storing lipotoxic free fatty acids (FFAs) as inert TGs due to chronic perturbation in lipid metabolism and lipotoxic insults. However, when liver hepatocytes' adaptive mechanisms are overburdened, lipotoxicity occurs, contributing to reactive oxygen species (ROS) formation, mitochondrial dysfunction, and endoplasmic reticulum (ER) stress. Impaired mitochondrial fatty acid oxidation, reduction in mitochondrial quality, and disrupted mitochondrial function are associated with a decrease in the energy levels and impaired redox balance and negatively affect mitochondria hepatocyte tolerance towards damaging hits. However, the sequence of events underlying mitochondrial failure from steatosis to hepatocarcinoma is still yet to be fully clarified. This review provides an overview of our understanding of mitochondrial adaptation in initial NAFLD stages and highlights how hepatic mitochondrial dysfunction and heterogeneity contribute to disease pathophysiology progression, from steatosis to hepatocellular carcinoma. Improving our understanding of different aspects of hepatocytes' mitochondrial physiology in the context of disease development and progression is crucial to improving diagnosis, management, and therapy of NAFLD/NASH.
Collapse
Affiliation(s)
- Ricardo Amorim
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- CIQUP-IMS/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| | - Carina C Magalhães
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Fernanda Borges
- CIQUP-IMS/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| | - Paulo J Oliveira
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - José Teixeira
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| |
Collapse
|
24
|
Bellanti F, Lo Buglio A, Vendemiale G. Hepatic Mitochondria-Gut Microbiota Interactions in Metabolism-Associated Fatty Liver Disease. Metabolites 2023; 13:322. [PMID: 36984762 PMCID: PMC10057853 DOI: 10.3390/metabo13030322] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/17/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
The prevalence of metabolism-associated fatty liver disease (MAFLD) represents an urgent pandemic, complicated by a higher risk of morbidity and mortality as well as an increased socio-economic burden. There is growing evidence proving the impact of gut microbiota modifications on the development and progression of MAFLD through changes in metabolic pathways, modulation of the immune response, and activation of pro-inflammatory signals. Concurrently, metabolites produced by gut microbiota consisting of short chain fatty acids and bile acids contribute to the regulation of hepatic homeostasis by interacting with mitochondria. Evolving research indicates that innovative therapeutic targets for MAFLD may focus on gut microbiota-mitochondria interplay to regulate hepatic homeostasis. Recent investigations have explored the potential of new treatment strategies, such as prebiotics, probiotics, and metabolites, to change the composition of gut microbiota and simultaneously exert a positive impact on mitochondrial function to improve MAFLD. This review summarizes the significance of mitochondria and reports modifications in the composition of gut microbiota and its metabolites in MAFLD in order to illustrate the fascinating interplay between liver mitochondria and intestinal microbiota, discussing the potential effects of innovative treatments to modulate gut microbiota.
Collapse
Affiliation(s)
- Francesco Bellanti
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | | | | |
Collapse
|
25
|
Mollet IG, Macedo MP. Pre-Diabetes-Linked miRNA miR-193b-3p Targets PPARGC1A, Disrupts Metabolic Gene Expression Profile and Increases Lipid Accumulation in Hepatocytes: Relevance for MAFLD. Int J Mol Sci 2023; 24:ijms24043875. [PMID: 36835287 PMCID: PMC9965679 DOI: 10.3390/ijms24043875] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/06/2023] [Accepted: 02/11/2023] [Indexed: 02/17/2023] Open
Abstract
Distinct plasma microRNA profiles associate with different disease features and could be used to personalize diagnostics. Elevated plasma microRNA hsa-miR-193b-3p has been reported in patients with pre-diabetes where early asymptomatic liver dysmetabolism plays a crucial role. In this study, we propose the hypothesis that elevated plasma hsa-miR-193b-3p conditions hepatocyte metabolic functions contributing to fatty liver disease. We show that hsa-miR-193b-3p specifically targets the mRNA of its predicted target PPARGC1A/PGC1α and consistently reduces its expression in both normal and hyperglycemic conditions. PPARGC1A/PGC1α is a central co-activator of transcriptional cascades that regulate several interconnected pathways, including mitochondrial function together with glucose and lipid metabolism. Profiling gene expression of a metabolic panel in response to overexpression of microRNA hsa-miR-193b-3p revealed significant changes in the cellular metabolic gene expression profile, including lower expression of MTTP, MLXIPL/ChREBP, CD36, YWHAZ and GPT, and higher expression of LDLR, ACOX1, TRIB1 and PC. Overexpression of hsa-miR-193b-3p under hyperglycemia also resulted in excess accumulation of intracellular lipid droplets in HepG2 cells. This study supports further research into potential use of microRNA hsa-miR-193b-3p as a possible clinically relevant plasma biomarker for metabolic-associated fatty liver disease (MAFLD) in dysglycemic context.
Collapse
Affiliation(s)
- Inês Guerra Mollet
- iNOVA4Health, NOVA Medical School (NMS), Faculdade de Ciências Médicas (FCM), Universidade NOVA de Lisboa, 1150-082 Lisboa, Portugal
- UCIBIO-Requimte, Faculdade de Ciências e Tecnologia (FCT), Universidade NOVA de Lisboa, 2825-149 Caparica, Portugal
- Correspondence: (I.G.M.); (M.P.M.)
| | - Maria Paula Macedo
- iNOVA4Health, NOVA Medical School (NMS), Faculdade de Ciências Médicas (FCM), Universidade NOVA de Lisboa, 1150-082 Lisboa, Portugal
- Associação Protectora dos Diabéticos de Portugal, Education Research Center (APDP-ERC), 1250-203 Lisbon, Portugal
- Correspondence: (I.G.M.); (M.P.M.)
| |
Collapse
|
26
|
Ramatchandirin B, Pearah A, He L. Regulation of Liver Glucose and Lipid Metabolism by Transcriptional Factors and Coactivators. Life (Basel) 2023; 13:life13020515. [PMID: 36836874 PMCID: PMC9962321 DOI: 10.3390/life13020515] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023] Open
Abstract
The prevalence of nonalcoholic fatty liver disease (NAFLD) worldwide is on the rise and NAFLD is becoming the most common cause of chronic liver disease. In the USA, NAFLD affects over 30% of the population, with similar occurrence rates reported from Europe and Asia. This is due to the global increase in obesity and type 2 diabetes mellitus (T2DM) because patients with obesity and T2DM commonly have NAFLD, and patients with NAFLD are often obese and have T2DM with insulin resistance and dyslipidemia as well as hypertriglyceridemia. Excessive accumulation of triglycerides is a hallmark of NAFLD and NAFLD is now recognized as the liver disease component of metabolic syndrome. Liver glucose and lipid metabolisms are intertwined and carbon flux can be used to generate glucose or lipids; therefore, in this review we discuss the important transcription factors and coactivators that regulate glucose and lipid metabolism.
Collapse
Affiliation(s)
| | - Alexia Pearah
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ling He
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, 600 N. Wolfe St, Baltimore, MD 21287, USA
- Correspondence: ; Tel.: +1-410-502-5765; Fax: +1-410-502-5779
| |
Collapse
|
27
|
Stevanović-Silva J, Beleza J, Coxito P, Oliveira PJ, Ascensão A, Magalhães J. Gestational Exercise Antagonises the Impact of Maternal High-Fat High-Sucrose Diet on Liver Mitochondrial Alterations and Quality Control Signalling in Male Offspring. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:1388. [PMID: 36674144 PMCID: PMC9858977 DOI: 10.3390/ijerph20021388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/03/2023] [Accepted: 01/09/2023] [Indexed: 06/17/2023]
Abstract
Maternal high-caloric nutrition and related gestational diabetes mellitus (GDM) are relevant modulators of the intrauterine environment, increasing the risk of liver metabolic alterations in mothers and offspring. In contrast, as a non-pharmacological approach against metabolic disorders, exercise is highly recommended in GDM treatment. We analysed whether gestational exercise (GE) protects mothers from diet-induced GDM metabolic consequences and mitigates liver mitochondrial deleterious alterations in their 6-week-old male offspring. Female Sprague Dawley rats were fed with control or high-fat high-sucrose (HFHS) diet and kept sedentary or submitted to GE. Male offspring were sedentary and fed with control diet. Sedentary HFHS mothers and their offspring showed impaired hepatic mitochondrial biogenesis and morphological evidence of mitochondrial remodelling. In contrast, GE-related beneficial effects were demonstrated by upregulation of mitochondrial biogenesis signalling markers and mitochondrial fusion proteins and downregulation of mitochondrial fission protein. Alterations in miR-34a, miR-130b, and miR-494, associated with epigenetic regulation of mitochondrial biogenesis, suggested that GE is a more critical modulator of intergenerational changes in miRs expression than the maternal diet. Our data showed that GE positively modulated the altered hepatic mitochondrial biogenesis and dynamics markers and quality control signalling associated with maternal HFHS-diet-related GDM in mothers and offspring.
Collapse
Affiliation(s)
- Jelena Stevanović-Silva
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sport, University of Porto, 4200-450 Porto, Portugal
| | - Jorge Beleza
- Department of Cell Biology, Physiology & Immunology, Faculty of Biology, University of Barcelona, 08028 Barcelona, Spain
| | - Pedro Coxito
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sport, University of Porto, 4200-450 Porto, Portugal
| | - Paulo J. Oliveira
- CNC—Center for Neuroscience and Cell Biology, CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - António Ascensão
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sport, University of Porto, 4200-450 Porto, Portugal
| | - José Magalhães
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sport, University of Porto, 4200-450 Porto, Portugal
| |
Collapse
|
28
|
Diclofenac Disrupts the Circadian Clock and through Complex Cross-Talks Aggravates Immune-Mediated Liver Injury-A Repeated Dose Study in Minipigs for 28 Days. Int J Mol Sci 2023; 24:ijms24021445. [PMID: 36674967 PMCID: PMC9863319 DOI: 10.3390/ijms24021445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/28/2022] [Accepted: 12/30/2022] [Indexed: 01/14/2023] Open
Abstract
Diclofenac effectively reduces pain and inflammation; however, its use is associated with hepato- and nephrotoxicity. To delineate mechanisms of injury, we investigated a clinically relevant (3 mg/kg) and high-dose (15 mg/kg) in minipigs for 4 weeks. Initially, serum biochemistries and blood-smears indicated an inflammatory response but returned to normal after 4 weeks of treatment. Notwithstanding, histopathology revealed drug-induced hepatitis, marked glycogen depletion, necrosis and steatosis. Strikingly, the genomic study revealed diclofenac to desynchronize the liver clock with manifest inductions of its components CLOCK, NPAS2 and BMAL1. The > 4-fold induced CRY1 expression underscored an activated core-loop, and the dose dependent > 60% reduction in PER2mRNA repressed the negative feedback loop; however, it exacerbated hepatotoxicity. Bioinformatics enabled the construction of gene-regulatory networks, and we linked the disruption of the liver-clock to impaired glycogenesis, lipid metabolism and the control of immune responses, as shown by the 3-, 6- and 8-fold induced expression of pro-inflammatory CXCL2, lysozyme and ß-defensin. Additionally, diclofenac treatment caused adrenocortical hypertrophy and thymic atrophy, and we evidenced induced glucocorticoid receptor (GR) activity by immunohistochemistry. Given that REV-ERB connects the circadian clock with hepatic GR, its > 80% repression alleviated immune responses as manifested by repressed expressions of CXCL9(90%), CCL8(60%) and RSAD2(70%). Together, we propose a circuitry, whereby diclofenac desynchronizes the liver clock in the control of the hepatic metabolism and immune response.
Collapse
|
29
|
Ma X, Ding WX. Methods for Monitoring Mitochondrial Biogenesis and Turnover in Cultured Hepatocytes and Mouse Liver Using MitoTimer Reporter Assay. Methods Mol Biol 2023; 2675:97-107. [PMID: 37258758 DOI: 10.1007/978-1-0716-3247-5_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Mitochondrial biogenesis and turnover rate are critical to maintain homeostasis of the intracellular mitochondrial pool. Altered mitochondrial biogenesis and mitophagy are closely related to many chronic diseases, highlighting the importance of mitochondrial stasis in various pathological conditions including liver diseases. We describe a detailed protocol for monitoring mitochondrial lifecycle in primary cultured mouse hepatocytes and mouse liver using the dual color fluorescence-based imaging of MitoTimer. Three types of mitochondria were visualized in mouse hepatocytes: green-only mitochondria (newly synthesized mitochondria), red-only mitochondria (old/aging mitochondria), as well as the majority of yellow mitochondria (representing an intermediate stage of mitochondria). The ratio of red/green fluorescence in each cell will be used to track mitochondrial aging. Super-resolution microscopy analysis revealed that majority of mitochondria were spatially heterogeneous with proteins from simultaneous new synthesis, maturation, and turnover in hepatocytes. MitoTimer reporter assay can specifically target to mitochondria and be used to monitor mitochondrial biogenesis and maturation as well as turnover in vitro and in vivo.
Collapse
Affiliation(s)
- Xiaowen Ma
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS, USA
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
30
|
Clare K, Dillon JF, Brennan PN. Reactive Oxygen Species and Oxidative Stress in the Pathogenesis of MAFLD. J Clin Transl Hepatol 2022; 10:939-946. [PMID: 36304513 PMCID: PMC9547261 DOI: 10.14218/jcth.2022.00067] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/22/2022] [Accepted: 06/06/2022] [Indexed: 12/04/2022] Open
Abstract
The pathogenesis of metabolic-associated fatty liver disease (MAFLD) is complex and thought to be dependent on multiple parallel hits on a background of genetic susceptibility. The evidence suggests that MAFLD progression is a dynamic two-way process relating to repetitive bouts of metabolic stress and inflammation interspersed with endogenous anti-inflammatory reparative responses. In MAFLD, excessive hepatic lipid accumulation causes the production of lipotoxins that induce mitochondrial dysfunction, endoplasmic reticular stress, and over production of reactive oxygen species (ROS). Models of MAFLD show marked disruption of mitochondrial function and reduced oxidative capacitance with impact on cellular processes including mitophagy, oxidative phosphorylation, and mitochondrial biogenesis. In excess, ROS modify insulin and innate immune signaling and alter the expression and activity of essential enzymes involved in lipid homeostasis. ROS can also cause direct damage to intracellular structures causing hepatocyte injury and death. In select cases, the use of anti-oxidants and ROS scavengers have been shown to diminish the pro-apoptopic effects of fatty acids. Given this link, endogenous anti-oxidant pathways have been a target of interest, with Nrf2 activation showing a reduction in oxidative stress and inflammation in models of MAFLD. Thyroid hormone receptor β (THRβ) agonists and nuclear peroxisome proliferation-activated receptor (PPAR) family have also gained interest in reducing hepatic lipotoxicity and restoring hepatic function in models of MAFLD. Unfortunately, the true interplay between the clinical and molecular components of MAFLD progression remain only partly understood. Most recently, multiomics-based strategies are being adopted for hypothesis-free analysis of the molecular changes in MAFLD. Transcriptome profiling maps the unique genotype-phenotype associations in MAFLD and with various single-cell transcriptome-based projects underway, there is hope of novel physiological insights to MAFLD progression and uncover therapeutic targets.
Collapse
Affiliation(s)
- Kathleen Clare
- Royal Alexandra Hospital, Paisley, NHS Greater Glasgow and Clyde, PA2 9PN, UK
| | - John F. Dillon
- University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK
| | - Paul N. Brennan
- University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK
- University of Edinburgh, Edinburgh BioQuarter, 5 Little France Drive, EH16 4UU, UK
- Correspondence to: Paul N. Brennan, University of Dundee, Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK. ORCID: https://orcid.org/0000-0001-8368-1478. Tel: +44-7445308786, E-mail:
| |
Collapse
|
31
|
Weber AA, Yang X, Mennillo E, Ding J, Watrous JD, Jain M, Chen S, Karin M, Tukey RH. Lactational delivery of Triclosan promotes non-alcoholic fatty liver disease in newborn mice. Nat Commun 2022; 13:4346. [PMID: 35896521 PMCID: PMC9329322 DOI: 10.1038/s41467-022-31947-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 07/11/2022] [Indexed: 11/21/2022] Open
Abstract
Here we show that Triclosan (TCS), a high-volume antimicrobial additive that has been detected in human breastmilk, can be efficiently transferred by lactation to newborn mice, causing significant fatty liver (FL) during the suckling period. These findings are relevant since pediatric non-alcoholic fatty liver disease (NAFLD) is escalating in the United States, with a limited mechanistic understanding. Lactational delivery stimulated hepatosteatosis, triglyceride accumulation, endoplasmic reticulum (ER) stress, signs of inflammation, and liver fibrosis. De novo lipogenesis (DNL) induced by lactational TCS exposure is shown to be mediated in a PERK-eIF2α-ATF4-PPARα cascade. The administration of obeticholic acid (OCA), a potent FXR agonist, as well as activation of intestinal mucosal-regenerative gp130 signaling, led to reduced liver ATF4 expression, PPARα signaling, and DNL when neonates were exposed to TCS. It is yet to be investigated but mother to child transmission of TCS or similar toxicants may underlie the recent increases in pediatric NAFLD.
Collapse
Affiliation(s)
- André A Weber
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Xiaojing Yang
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Elvira Mennillo
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Jeffrey Ding
- Departments of Medicine and Pharmacology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Jeramie D Watrous
- Departments of Medicine and Pharmacology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Mohit Jain
- Departments of Medicine and Pharmacology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Shujuan Chen
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Robert H Tukey
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California, San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
32
|
Wang M, Zhu Z, Kan Y, Yu M, Guo W, Ju M, Wang J, Yi S, Han S, Shang W, Zhang Z, Zhang L, Fang P. Treatment with spexin mitigates diet-induced hepatic steatosis in vivo and in vitro through activation of galanin receptor 2. Mol Cell Endocrinol 2022; 552:111688. [PMID: 35654225 DOI: 10.1016/j.mce.2022.111688] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 04/28/2022] [Accepted: 05/25/2022] [Indexed: 01/12/2023]
Abstract
It was reported that spexin as an adipocyte-secreted protein could regulate obesity and insulin resistance. However, the specific metabolic contribution of spexin to fatty liver remains incompletely understood. Herein, we investigated the effects of spexin on hepatosteatosis and explored the underlying molecular mechanisms. HFD-fed mice were injected with spexin and/or GALR2 antagonist M871, while PA-induced HepG2 cells were treated with spexin in the absence or presence of M871 for 12 h, respectively. Gene expression in liver tissues and hepatocytes was assessed by qRT-PCR and western blotting, respectively. The results showed that body weight, visceral fat content, liver lipid droplet formation, hepatic intracellular triglyceride, and serum triglyceride were reduced in spexin-treated mice. Furthermore, spexin increased the expression of hepatic CPT1A, PPARα, SIRT1, KLF9, PGC-1α and PEPCK in vivo and in vitro. Additionally, spexin treatment improved glucose tolerance and insulin sensitivity in mice fed the HFD. Interestingly, these spexin-mediated beneficial effects were abolished by the GALR2 antagonist M871 in mice fed HFD and PA-induced HepG2 cells, suggesting that spexin mitigated HFD-induced hepatic steatosis by activating the GALR2, thereby increasing CPT1A, PPARα, SIRT1, KLF9, PGC-1α and PEPCK expression. Taken together, these data suggest that spexin ameliorates NAFLD by improving lipolysis and fatty acid oxidation via activation of GALR2 signaling.
Collapse
Affiliation(s)
- Mengyuan Wang
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ziyue Zhu
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yue Kan
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Mei Yu
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Wancheng Guo
- Department of Endocrinology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, 225001, China
| | - Mengxian Ju
- Department of Endocrinology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, 225001, China
| | - Junjun Wang
- Department of Endocrinology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, 225001, China
| | - Shuxin Yi
- Department of Endocrinology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, 225001, China
| | - Shiyu Han
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Wenbin Shang
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhenwen Zhang
- Department of Endocrinology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, 225001, China.
| | - Li Zhang
- Hanlin College, Nanjing University of Chinese Medicine, Taizhou, 225300, China.
| | - Penghua Fang
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Hanlin College, Nanjing University of Chinese Medicine, Taizhou, 225300, China.
| |
Collapse
|
33
|
Cook JJ, Wei M, Segovia B, Cosio-Lima L, Simpson J, Taylor S, Koh Y, Kim S, Lee Y. Endurance exercise-mediated metabolic reshuffle attenuates high-caloric diet-induced non-alcoholic fatty liver disease. Ann Hepatol 2022; 27:100709. [PMID: 35489641 DOI: 10.1016/j.aohep.2022.100709] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/17/2022] [Accepted: 04/04/2022] [Indexed: 02/04/2023]
Abstract
INTRODUCTION AND AIM Non-alcoholic fatty liver disease (NAFLD) is one of the most common diseases in the United States. Metabolic distress (obese diabetes) is the main causative element of NAFLD. While there is no cure for NAFLD, endurance exercise (EEx) has emerged as a therapeutic strategy against NAFLD. However, mechanisms of EXE-induced hepatic protection especially in female subjects remain unidentified. Thus, the aim of the study is to examine molecular mechanisms of EXE-induced hepatic protection against diet-induced NAFLD in female mice. MATERIAL AND METHODS Nine-week-old female C57BL/6J mice were randomly divided into three groups: normal-diet control group (CON, n=11); high-fat diet/high-fructose group (HFD/HF, n=11); and HFD/HF+EEx group (HFD/HF+EEx, n=11). The mice assigned to HFD/HF and HFD/HF+EEx groups were fed with HFD/HF for 12 weeks, after which the mice assigned to the EEx group began treadmill exercise for 12 weeks, with HFD/HF continued. RESULTS EEx attenuated hepatic steatosis, reduced de novo lipogenesis (reduction in ATP-Citrate- Lyase and diacylglycerol-O-acyltransferase 1), and enhanced mitochondrial biogenesis and fatty-acid activation (oxidative phosphorylation enzymes and Acyl-CoA synthetase1). Also, EEx prevented upregulation of gluconeogenic proteins (glyceraldehyde-3-phosphate dehydrogenase, glucose-6-phosphatase, and phosphoenolpyruvate-carboxykinase1), premature senescence (suppression of p53, p22, and p16, tumor-necrosis-factor-α, and interleukin-1β, and oxidative stress), and autophagy deficiency. Furthermore, EXE reversed apoptosis arrest (cleaved cysteine-dependent-aspartate-directed protease3 and Poly-(ADP-ribose)-polymerase1). CONCLUSION EEx-mediated reparations of metabolic and redox imbalance (utilization of pentose phosphate pathway), and autophagy deficiency caused by metabolic distress critically contribute to preventing/delaying severe progression of NAFLD. Also, EEx-induced anti-senescence and cell turnover are crucial protective mechanisms against NAFLD.
Collapse
Affiliation(s)
- Joshua J Cook
- Molecular and Cellular Exercise Physiology Laboratory, Department of Movement Sciences and Health, Usha Kundu, MD College of Health, University of West Florida, Pensacola, FL 32514, USA
| | - Madeline Wei
- Molecular and Cellular Exercise Physiology Laboratory, Department of Movement Sciences and Health, Usha Kundu, MD College of Health, University of West Florida, Pensacola, FL 32514, USA
| | - Benny Segovia
- Molecular and Cellular Exercise Physiology Laboratory, Department of Movement Sciences and Health, Usha Kundu, MD College of Health, University of West Florida, Pensacola, FL 32514, USA
| | - Ludmila Cosio-Lima
- Molecular and Cellular Exercise Physiology Laboratory, Department of Movement Sciences and Health, Usha Kundu, MD College of Health, University of West Florida, Pensacola, FL 32514, USA
| | - Jeffrey Simpson
- Molecular and Cellular Exercise Physiology Laboratory, Department of Movement Sciences and Health, Usha Kundu, MD College of Health, University of West Florida, Pensacola, FL 32514, USA
| | - Scott Taylor
- Department of Biology, Hal Marcus College of Science and Engineering, University of West Florida, Pensacola, FL 32514, USA
| | - Yunsuk Koh
- Department of Health, Human Performance and Recreation, Robbins College of Human Sciences, Baylor University, Waco, TX 76798, USA
| | - Sangho Kim
- Department of Sport Science, College of Culture and Sports, School of Global Sport Studies, Korea University, Sejong 30019, South Korea
| | - Youngil Lee
- Molecular and Cellular Exercise Physiology Laboratory, Department of Movement Sciences and Health, Usha Kundu, MD College of Health, University of West Florida, Pensacola, FL 32514, USA.
| |
Collapse
|
34
|
Ramanathan R, Ali AH, Ibdah JA. Mitochondrial Dysfunction Plays Central Role in Nonalcoholic Fatty Liver Disease. Int J Mol Sci 2022; 23:ijms23137280. [PMID: 35806284 PMCID: PMC9267060 DOI: 10.3390/ijms23137280] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 12/04/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a global pandemic that affects one-quarter of the world’s population. NAFLD includes a spectrum of progressive liver disease from steatosis to nonalcoholic steatohepatitis (NASH), fibrosis, and cirrhosis and can be complicated by hepatocellular carcinoma. It is strongly associated with metabolic syndromes, obesity, and type 2 diabetes, and it has been shown that metabolic dysregulation is central to its pathogenesis. Recently, it has been suggested that metabolic- (dysfunction) associated fatty liver disease (MAFLD) is a more appropriate term to describe the disease than NAFLD, which puts increased emphasis on the important role of metabolic dysfunction in its pathogenesis. There is strong evidence that mitochondrial dysfunction plays a significant role in the development and progression of NAFLD. Impaired mitochondrial fatty acid oxidation and, more recently, a reduction in mitochondrial quality, have been suggested to play a major role in NAFLD development and progression. In this review, we provide an overview of our current understanding of NAFLD and highlight how mitochondrial dysfunction contributes to its pathogenesis in both animal models and human subjects. Further we discuss evidence that the modification of mitochondrial function modulates NAFLD and that targeting mitochondria is a promising new avenue for drug development to treat NAFLD/NASH.
Collapse
Affiliation(s)
- Raghu Ramanathan
- Division of Gastroenterology and Hepatology, University of Missouri, Columbia, MO 65212, USA; (R.R.); (A.H.A.)
- Harry S. Truman Memorial Veterans Medical Center, Columbia, MO 65201, USA
| | - Ahmad Hassan Ali
- Division of Gastroenterology and Hepatology, University of Missouri, Columbia, MO 65212, USA; (R.R.); (A.H.A.)
- Harry S. Truman Memorial Veterans Medical Center, Columbia, MO 65201, USA
| | - Jamal A. Ibdah
- Division of Gastroenterology and Hepatology, University of Missouri, Columbia, MO 65212, USA; (R.R.); (A.H.A.)
- Harry S. Truman Memorial Veterans Medical Center, Columbia, MO 65201, USA
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO 65212, USA
- Correspondence: ; Tel.: +573-882-7349; Fax: +573-884-4595
| |
Collapse
|
35
|
Grieb BC, Eischen CM. MTBP and MYC: A Dynamic Duo in Proliferation, Cancer, and Aging. BIOLOGY 2022; 11:881. [PMID: 35741402 PMCID: PMC9219613 DOI: 10.3390/biology11060881] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 05/29/2022] [Accepted: 06/02/2022] [Indexed: 12/21/2022]
Abstract
The oncogenic transcription factor c-MYC (MYC) is highly conserved across species and is frequently overexpressed or dysregulated in human cancers. MYC regulates a wide range of critical cellular and oncogenic activities including proliferation, metabolism, metastasis, apoptosis, and differentiation by transcriptionally activating or repressing the expression of a large number of genes. This activity of MYC is not carried out in isolation, instead relying on its association with a myriad of protein cofactors. We determined that MDM Two Binding Protein (MTBP) indirectly binds MYC and is a novel MYC transcriptional cofactor. MTBP promotes MYC-mediated transcriptional activity, proliferation, and cellular transformation by binding in a protein complex with MYC at MYC-bound promoters. This discovery provided critical context for data linking MTBP to aging as well as a rapidly expanding body of evidence demonstrating MTBP is overexpressed in many human malignancies, is often linked to poor patient outcomes, and is necessary for cancer cell survival. As such, MTBP represents a novel and potentially broad reaching oncologic drug target, particularly when MYC is dysregulated. Here we have reviewed the discovery of MTBP and the initial controversy with its function as well as its associations with proliferation, MYC, DNA replication, aging, and human cancer.
Collapse
Affiliation(s)
- Brian C. Grieb
- Vanderbilt-Ingram Cancer Center, Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
- Department of Cell & Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Christine M. Eischen
- Department of Cancer Biology and the Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
36
|
Miao H, Ouyang H, Guo Q, Wei M, Lu B, Kai G, Ji L. Chlorogenic acid alleviated liver fibrosis in methionine and choline deficient diet-induced nonalcoholic steatohepatitis in mice and its mechanism. J Nutr Biochem 2022; 106:109020. [PMID: 35472433 DOI: 10.1016/j.jnutbio.2022.109020] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 01/22/2022] [Accepted: 03/21/2022] [Indexed: 01/19/2023]
Abstract
Nonalcoholic steatohepatitis (NASH), one of the most common chronic liver diseases, is a progressive form of nonalcoholic fatty liver disease (NAFLD) accompanied by the development of liver fibrosis. Chlorogenic acid (CGA) is a natural polyphenolic compound. This study aims to observe the CGA-provided alleviation on liver fibrosis in methionine and choline deficient (MCD) diet-induced NASH in mice and to elucidate its engaged mechanism. CGA attenuated hepatocellular injury, decreased the elevated hepatic lipids accumulation and attenuated liver fibrosis by reducing hepatic collagen deposition in mice fed with MCD diet. CGA abrogated the activation of hepatic stellate cells (HSCs) and promoted mitochondrial biogenesis both in vivo and in vitro. Moreover, the CGA-provided inhibition on HSCs activation in vitro was obviously disappeared after the application of peroxisome proliferator-activated receptor gamma, coactivator 1alpha (PGC1α) siRNA. CGA reduced the enhanced hepatic extracellular matrix (ECM) expression and the elevated serum high-mobility group box 1 (HMGB1) content in mice fed with MCD diet. CGA decreased the HMGB1-induced ECM production in both human liver sinusoidal endothelial cells (LSECs) and human umbilical vein endothelial cells (HUVECs). CGA also weakly promoted mitochondrial biogenesis in both LSECs and HUVECs incubated with HMGB1. Hence, CGA ameliorated hepatic fibrosis in mice fed with MCD diet through inhibiting HSCs activation via promoting mitochondrial biogenesis and reducing the HMGB1-initiated ECM production in hepatic vascular endothelial cells.
Collapse
Affiliation(s)
- Hui Miao
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hao Ouyang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qian Guo
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Mengjuan Wei
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Bin Lu
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Guoyin Kai
- Laboratory of Medicinal Plant Biotechnology, College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 311402, China.
| | - Lili Ji
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
37
|
Jiang JJ, Zhang GF, Zheng JY, Sun JH, Ding SB. Targeting Mitochondrial ROS-Mediated Ferroptosis by Quercetin Alleviates High-Fat Diet-Induced Hepatic Lipotoxicity. Front Pharmacol 2022; 13:876550. [PMID: 35496312 PMCID: PMC9039018 DOI: 10.3389/fphar.2022.876550] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 03/24/2022] [Indexed: 12/31/2022] Open
Abstract
Background: The protective effect of quercetin on nonalcoholic fatty liver disease (NAFLD) has been reported, but its mechanism remains poorly understood. Recently, quercetin was reported to be capable of inhibiting ferroptosis, which is a recognized type of regulated cell death. Moreover, hepatic ferroptosis plays an important role in the progression of NAFLD, but experimental evidence is limited. Hence, our study aimed to investigate the effect of quercetin on hepatic ferroptosis in high-fat diet (HFD)-induced NAFLD and further elucidate the underlying molecular mechanism. Methods: C57BL/6J mice were fed either a normal diet (ND), an HFD, or an HFD supplemented with quercetin for 12 weeks. Hepatic lipid peroxidation, steatosis, ferroptosis and iron overload were examined. In vitro, steatotic L-02 cells was used to study the potential mechanism. Results: We found that the HFD caused lipid peroxidation, lipid accumulation and ferroptosis in the liver, which were rescued by quercetin supplementation. Consistent with the in vivo results, quercetin alleviated lipid droplet accumulation and reduced the levels of lipid reactive oxygen species (ROS) and ferroptosis in steatotic L-02 cells. Using a mitochondrial ROS (MtROS) scavenger (Mito-TEMPO) and ferroptosis specific inhibitor (Fer-1), we found that quercetin remarkably alleviated lipid droplet accumulation and lipid peroxidation by reducing MtROS-mediated ferroptosis in steatotic L-02 cells. Conclusion: Our data showed that HFD consumption induced lipid accumulation and triggered ferroptosis in liver, ultimately leading to hepatic lipotoxicity, which can be alleviated by quercetin. Findings from this study provide new insight into the mechanism by which quercetin can be used for the prevention and treatment of NAFLD.
Collapse
Affiliation(s)
- Jin-Jin Jiang
- Jiangsu Vocational College of Medicine, Yancheng, China
| | - Guo-Fu Zhang
- Department of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Jia-Yi Zheng
- Jiangsu Vocational College of Medicine, Yancheng, China
| | - Ji-Hu Sun
- Jiangsu Vocational College of Medicine, Yancheng, China
- *Correspondence: Shi-Bin Ding, ; Ji-Hu Sun,
| | - Shi-Bin Ding
- Jiangsu Vocational College of Medicine, Yancheng, China
- *Correspondence: Shi-Bin Ding, ; Ji-Hu Sun,
| |
Collapse
|
38
|
Hepatocyte-specific activity of TSC22D4 triggers progressive NAFLD by impairing mitochondrial function. Mol Metab 2022; 60:101487. [PMID: 35378329 PMCID: PMC9034319 DOI: 10.1016/j.molmet.2022.101487] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/28/2022] [Accepted: 03/28/2022] [Indexed: 11/24/2022] Open
Abstract
Objective Fibrotic organ responses have recently been identified as long-term complications in diabetes. Indeed, insulin resistance and aberrant hepatic lipid accumulation represent driving features of progressive non-alcoholic fatty liver disease (NAFLD), ranging from simple steatosis and non-alcoholic steatohepatitis (NASH) to fibrosis. Effective pharmacological regimens to stop progressive liver disease are still lacking to-date. Methods Based on our previous discovery of transforming growth factor beta-like stimulated clone (TSC)22D4 as a key driver of insulin resistance and glucose intolerance in obesity and type 2 diabetes, we generated a TSC22D4-hepatocyte specific knockout line (TSC22D4-HepaKO) and exposed mice to control or NASH diet models. Mechanistic insights were generated by metabolic phenotyping and single-nuclei RNA sequencing. Results Hepatic TSC22D4 expression was significantly correlated with markers of liver disease progression and fibrosis in both murine and human livers. Indeed, hepatic TSC22D4 levels were elevated in human NASH patients as well as in several murine NASH models. Specific genetic deletion of TSC22D4 in hepatocytes led to reduced liver lipid accumulation, improvements in steatosis and inflammation scores and decreased apoptosis in mice fed a lipogenic MCD diet. Single-nuclei RNA sequencing revealed a distinct TSC22D4-dependent gene signature identifying an upregulation of mitochondrial-related processes in hepatocytes upon loss of TSC22D4. An enrichment of genes involved in the TCA cycle, mitochondrial organization, and triglyceride metabolism underscored the hepatocyte-protective phenotype and overall decreased liver damage as seen in mouse models of hepatocyte-selective TSC22D4 loss-of-function. Conclusions Together, our data uncover a new connection between targeted depletion of TSC22D4 and intrinsic metabolic processes in progressive liver disease. Hepatocyte-specific reduction of TSC22D4 improves hepatic steatosis and promotes hepatocyte survival via mitochondrial-related mechanisms thus paving the way for targeted therapies. TSC22D4 is significantly upregulated in both patients with NASH and liver fibrosis and in murine models of NAFLD. Hepatocyte-specific knockdown of TSC22D4 results in reduced steatosis and hepatocyte damage in MCD diet fed mice. Single-nuclei RNA sequencing from TSC22D4-HepaKO mice reveals enrichment in lipid metabolism and OXPHOS pathways. Loss of TSC22D4 increases the oxidative consumption rate and upregulates mitochondrial maintenance genes in hepatocytes. TSC22D4 expression negatively correlates with mitochondrial targets in NASH patients.
Collapse
|
39
|
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
|
40
|
Mooli RGR, Mukhi D, Ramakrishnan SK. Oxidative Stress and Redox Signaling in the Pathophysiology of Liver Diseases. Compr Physiol 2022; 12:3167-3192. [PMID: 35578969 PMCID: PMC10074426 DOI: 10.1002/cphy.c200021] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The increased production of derivatives of molecular oxygen and nitrogen in the form of reactive oxygen species (ROS) and reactive nitrogen species (RNS) lead to molecular damage called oxidative stress. Under normal physiological conditions, the ROS generation is tightly regulated in different cells and cellular compartments. Any disturbance in the balance between the cellular generation of ROS and antioxidant balance leads to oxidative stress. In this article, we discuss the sources of ROS (endogenous and exogenous) and antioxidant mechanisms. We also focus on the pathophysiological significance of oxidative stress in various cell types of the liver. Oxidative stress is implicated in the development and progression of various liver diseases. We narrate the master regulators of ROS-mediated signaling and their contribution to liver diseases. Nonalcoholic fatty liver diseases (NAFLD) are influenced by a "multiple parallel-hit model" in which oxidative stress plays a central role. We highlight the recent findings on the role of oxidative stress in the spectrum of NAFLD, including fibrosis and liver cancer. Finally, we provide a brief overview of oxidative stress biomarkers and their therapeutic applications in various liver-related disorders. Overall, the article sheds light on the significance of oxidative stress in the pathophysiology of the liver. © 2022 American Physiological Society. Compr Physiol 12:3167-3192, 2022.
Collapse
Affiliation(s)
- Raja Gopal Reddy Mooli
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Dhanunjay Mukhi
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Sadeesh K Ramakrishnan
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
41
|
Stevanović-Silva J, Beleza J, Coxito P, Costa RC, Ascensão A, Magalhães J. Fit mothers for a healthy future: Breaking the intergenerational cycle of non-alcoholic fatty liver disease with maternal exercise. Eur J Clin Invest 2022; 52:e13596. [PMID: 34120338 DOI: 10.1111/eci.13596] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/01/2021] [Accepted: 05/06/2021] [Indexed: 12/20/2022]
Abstract
UNLABELLED SPECIAL ISSUE: 'FOIEGRAS-Bioenergetic Remodelling in the Pathophysiology and Treatment of Non-Alcoholic Fatty Liver Disease'. BACKGROUND Non-alcoholic fatty liver disease (NAFLD) emerges as significant health burden worldwide. Lifestyle changes, unhealthy dietary habits and physical inactivity, can trigger NAFLD development. Persisting on these habits during pregnancy affects in utero environment and prompts a specific metabolic response in foetus resulting in offspring metabolic maladjustments potentially critical for developing NAFLD later in life. The increasing prevalence of NAFLD, particularly in children, has shifted the research focus towards preventive and therapeutic strategies. Yet, designing effective approaches that can break the NAFLD intergenerational cycle becomes even more complicated. Regular physical exercise (PE) is a powerful non-pharmacological strategy known to counteract deleterious metabolic outcomes. In this narrative review, we aimed to briefly describe NAFLD pathogenesis focusing on maternal nutritional challenge and foetal programming, and to provide potential mechanisms behind the putative intergenerational effect of PE against metabolic diseases, including liver diseases. METHODS Following detailed electronic database search, recent existing evidence about NAFLD development, intergenerational programming and gestational exercise effects was critically analysed and discussed. RESULTS PE during pregnancy could have a great potential to counteract intergenerational transmission of metabolic burden. The interplay between different PE roles-metabolic, endocrine and epigenetic-could offer a more stable in utero environment to the foetus, thus rescuing offspring vulnerability to metabolic disturbances. CONCLUSIONS The better understanding of maternal PE beneficial consequences on offspring metabolism could reinforce the importance of PE during pregnancy as an indispensable strategy in improving offspring health.
Collapse
Affiliation(s)
- Jelena Stevanović-Silva
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sport, University of Porto, Porto, Portugal
| | - Jorge Beleza
- Department of Cell Biology, Physiology & Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Pedro Coxito
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sport, University of Porto, Porto, Portugal
| | - Rui Carlos Costa
- Department of Communication and Art, Research Institute for Design, Media and Culture (ID+), Aveiro University, Aveiro, Portugal
| | - António Ascensão
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sport, University of Porto, Porto, Portugal
| | - José Magalhães
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sport, University of Porto, Porto, Portugal
| |
Collapse
|
42
|
Khalil M, Rita Caponio G, Diab F, Shanmugam H, Di Ciaula A, Khalifeh H, Vergani L, Calasso M, De Angelis M, Portincasa P. Unraveling the beneficial effects of herbal Lebanese mixture “Za’atar”. History, studies, and properties of a potential healthy food ingredient. J Funct Foods 2022; 90:104993. [DOI: 10.1016/j.jff.2022.104993] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
43
|
Wang X, Xie Q. Metabolic Dysfunction-associated Fatty Liver Disease (MAFLD) and Viral Hepatitis. J Clin Transl Hepatol 2022; 10:128-133. [PMID: 35233381 PMCID: PMC8845159 DOI: 10.14218/jcth.2021.00200] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 08/19/2021] [Accepted: 09/07/2021] [Indexed: 12/04/2022] Open
Abstract
A new definition of metabolic dysfunction-associated fatty liver disease (MAFLD) was proposed in 2020. The change from nonalcoholic fatty liver disease (NAFLD) to MAFLD highlights the metabolic abnormalities that accompany fatty liver. The diagnosis of MAFLD does not require exclusion of secondary causes of liver diseases and alcohol consumption. Thus, MAFLD may coexist with other types of liver diseases, such as viral hepatitis, a disease that remains the most common cause of liver disease-related death. With the increasing prevalence of MAFLD, patients with coincidental MAFLD and viral hepatitis are frequently encountered in clinical practice. In this review, we mainly summarize the mutual relationship between hepatitis B/C and systematic metabolism dysfunction related to MAFLD. We discuss the impact of MAFLD on progression of viral hepatitis and the therapies. Some unaddressed clinical problems related to concomitant MAFLD and viral hepatitis are also identified.
Collapse
Affiliation(s)
- Xiaolin Wang
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qing Xie
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
44
|
Xu J, Chen S, Wang W, Man Lam S, Xu Y, Zhang S, Pan H, Liang J, Huang X, Wang Y, Li T, Jiang Y, Wang Y, Ding M, Shui G, Yang H, Huang X. Hepatic CDP-diacylglycerol synthase 2 deficiency causes mitochondrial dysfunction and promotes rapid progression of NASH and fibrosis. Sci Bull (Beijing) 2022; 67:299-314. [PMID: 36546079 DOI: 10.1016/j.scib.2021.10.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 09/23/2021] [Accepted: 10/14/2021] [Indexed: 01/06/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) encompasses a spectrum of pathologies, ranging from steatosis to nonalcoholic steatohepatitis (NASH). The factors promoting the progression of steatosis to NASH are still unclear. Recent studies suggest that mitochondrial lipid composition is critical in NASH development. Here, we showed that CDP-DAG synthase 2 (Cds2) was downregulated in genetic or diet-induced NAFLD mouse models. Liver-specific deficiency of Cds2 provoked hepatic steatosis, inflammation and fibrosis in five-week-old mice. CDS2 is enriched in mitochondria-associated membranes (MAMs), and hepatic Cds2 deficiency impaired mitochondrial function and decreased mitochondrial PE levels. Overexpression of phosphatidylserine decarboxylase (PISD) alleviated the NASH-like phenotype in Cds2f/f;AlbCre mice and abnormal mitochondrial morphology and function caused by CDS2 deficiency in hepatocytes. Additionally, dietary supplementation with an agonist of peroxisome proliferator-activated receptor alpha (PPARα) attenuated mitochondrial defects and ameliorated the NASH-like phenotype in Cds2f/f;AlbCre mice. Finally, Cds2 overexpression protected against high-fat diet-induced hepatic steatosis and obesity. Thus, Cds2 modulates mitochondrial function and NASH development.
Collapse
Affiliation(s)
- Jiesi Xu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Siyu Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wei Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Sin Man Lam
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yang Xu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Beijing 100050, China
| | - Shaohua Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Huimin Pan
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jingjing Liang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiahe Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yu Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Ting Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yuqiang Jiang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yingchun Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Mei Ding
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hongyuan Yang
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Xun Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
45
|
Xu S, Wang Y, Li Z, Hua Q, Jiang M, Fan X. LncRNA GAS5 Knockdown Mitigates Hepatic Lipid Accumulation via Regulating MiR-26a-5p/PDE4B to Activate cAMP/CREB Pathway. Front Endocrinol (Lausanne) 2022; 13:889858. [PMID: 35957809 PMCID: PMC9361042 DOI: 10.3389/fendo.2022.889858] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/23/2022] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE Non-alcoholic fatty liver disease (NAFLD) can be attributed to the dysregulation of hepatic lipid metabolism; however, its cellular and molecular mechanisms remain unclear. This study aims to explore the effect of long non-coding RNA growth arrest specific 5 (GAS5) on hepatic lipid metabolism in fatty liver models. METHODS Obese mice, high fat diet-fed mice and free fatty acid-stimulated cells were used for GAS5 expression detection. GAS5 overexpression or knockdown models were established to elucidate the regulatory function of GAS5 in de novo lipogenesis (DNL) and mitochondrial function. Bioinformatic analyses and dual luciferase assays were used to investigate the interaction between GAS5, miR-26a-5p and phosphodiesterase (PDE) 4B. The involvement of the cyclic adenosine monophosphate (cAMP)/cAMP-response element-binding protein (CREB) pathway was evaluated using H89 and forskolin treatment. RESULTS GAS5 was activated in vitro and in vivo fatty liver models. Knockdown of GAS5 reduced lipid droplet accumulation, DNL associated enzymes and preserved mitochondrial function, while GAS5 overexpression exacerbated hepatic lipid accumulation. Mechanistically, GAS5 sponged miR-26a-5p to increase PDE4B expression and subsequently modulated DNL and mitochondrial function via the cAMP/CREB pathway. CONCLUSION Downregulation of GAS5 can activate the cAMP/CREB pathway through miR-26a-5p/PDE4B axis to mitigate hepatic lipid accumulation. This study provides evidence that downregulation of GAS5 may be a potential therapeutic option for the treatment of NAFLD.
Collapse
Affiliation(s)
| | | | | | | | - Miao Jiang
- *Correspondence: Xiaoming Fan, ; Miao Jiang,
| | | |
Collapse
|
46
|
Roberts FL, Markby GR. New Insights into Molecular Mechanisms Mediating Adaptation to Exercise; A Review Focusing on Mitochondrial Biogenesis, Mitochondrial Function, Mitophagy and Autophagy. Cells 2021; 10:cells10102639. [PMID: 34685618 PMCID: PMC8533934 DOI: 10.3390/cells10102639] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/27/2021] [Accepted: 09/29/2021] [Indexed: 12/25/2022] Open
Abstract
Exercise itself is fundamental for good health, and when practiced regularly confers a myriad of metabolic benefits in a range of tissues. These benefits are mediated by a range of adaptive responses in a coordinated, multi-organ manner. The continued understanding of the molecular mechanisms of action which confer beneficial effects of exercise on the body will identify more specific pathways which can be manipulated by therapeutic intervention in order to prevent or treat various metabolism-associated diseases. This is particularly important as exercise is not an available option to all and so novel methods must be identified to confer the beneficial effects of exercise in a therapeutic manner. This review will focus on key emerging molecular mechanisms of mitochondrial biogenesis, autophagy and mitophagy in selected, highly metabolic tissues, describing their regulation and contribution to beneficial adaptations to exercise.
Collapse
|
47
|
Middleton P, Vergis N. Mitochondrial dysfunction and liver disease: role, relevance, and potential for therapeutic modulation. Therap Adv Gastroenterol 2021; 14:17562848211031394. [PMID: 34377148 PMCID: PMC8320552 DOI: 10.1177/17562848211031394] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 06/18/2021] [Indexed: 02/04/2023] Open
Abstract
Mitochondria are key organelles involved in energy production as well as numerous metabolic processes. There is a growing interest in the role of mitochondrial dysfunction in the pathogenesis of common chronic diseases as well as in cancer development. This review will examine the role mitochondria play in the pathophysiology of common liver diseases, including alcohol-related liver disease, non-alcoholic fatty liver disease, chronic hepatitis B and hepatocellular carcinoma. Mitochondrial dysfunction is described widely in the literature in studies examining patient tissue and in disease models. Despite significant differences in pathophysiology between chronic liver diseases, common mitochondrial defects are described, including increased mitochondrial reactive oxygen species production and impaired oxidative phosphorylation. We review the current literature on mitochondrial-targeted therapies, which have the potential to open new therapeutic avenues in the management of patients with chronic liver disease.
Collapse
Affiliation(s)
| | - Nikhil Vergis
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| |
Collapse
|
48
|
Losartan Prevents Hepatic Steatosis and Macrophage Polarization by Inhibiting HIF-1α in a Murine Model of NAFLD. Int J Mol Sci 2021; 22:ijms22157841. [PMID: 34360607 PMCID: PMC8346090 DOI: 10.3390/ijms22157841] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/13/2021] [Accepted: 07/20/2021] [Indexed: 12/21/2022] Open
Abstract
Hypoxia and hepatosteatosis microenvironments are fundamental traits of nonalcoholic fatty liver disease (NAFLD). Hypoxia-inducible factor-1α (HIF-1α) is a transcription factor that controls the cellular response to hypoxia and is activated in hepatocytes of patients with NAFLD, whereas the route and regulation of lipid droplets (LDs) and macrophage polarization related to systemic inflammation in NAFLD is unknown. Losartan is an angiotensin II receptor antagonist, that approved portal hypertension and related HIF-1α pathways in hepatic injury models. Here, we show that losartan in a murine model of NAFLD significantly decreased hepatic de novo lipogenesis (DNL) as well as suppressed lipid droplets (LDs), LD-associated proteins, perilipins (PLINs), and cell-death-inducing DNA-fragmentation-factor (DFF45)-like effector (CIDE) family in liver and epididymal white adipose tissues (EWAT) of ob/ob mice. Obesity-mediated macrophage M1 activation was also required for HIF-1α expression in the liver and EWAT of ob/ob mice. Administration of losartan significantly diminishes obesity-enhanced macrophage M1 activation and suppresses hepatosteatosis. Moreover, HIF-1α-mediated mitochondrial dysfunction was reversed in ob/ob mice treated with losartan. Together, the regulation of HIF-1α controls LDs protein expression and macrophage polarization, which highlights a potential target for losartan in NAFLD.
Collapse
|
49
|
Dusabimana T, Park EJ, Je J, Jeong K, Yun SP, Kim HJ, Kim H, Park SW. P2Y2R Deficiency Ameliorates Hepatic Steatosis by Reducing Lipogenesis and Enhancing Fatty Acid β-Oxidation through AMPK and PGC-1α Induction in High-Fat Diet-Fed Mice. Int J Mol Sci 2021; 22:ijms22115528. [PMID: 34073834 PMCID: PMC8197197 DOI: 10.3390/ijms22115528] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/10/2021] [Accepted: 05/18/2021] [Indexed: 12/23/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a chronic metabolic liver disease associated with obesity and insulin resistance. Activation of the purinergic receptor P2Y2R has been reported to promote adipogenesis, inflammation and dyslipidemia in adipose tissues in obese mice. However, the role of P2Y2R and its mechanisms in NAFLD remain unknown. We hypothesized that P2Y2R deficiency may play a protective role in NAFLD by modulating lipid metabolism in the liver. In this study, we fed wild type and P2Y2R knockout mice with a high-fat diet (HFD) for 12 weeks and analyzed metabolic phenotypes. First, P2Y2R deficiency effectively improved insulin resistance with a reduction in body weight and plasma insulin. Second, P2Y2R deficiency attenuated hepatic lipid accumulation and injury with reduced alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels. Third, P2Y2R deficiency decreased the expression of fatty acid synthesis mediators (cluster of differentiation (CD36), fatty acid synthase (FAS), and stearoyl-CoA desaturase 1 (SCD1)); and increased the expression of adipose triglyceride lipase (ATGL), a lipolytic enzyme. Mechanistically, P2Y2R deficiency increased the AMP-activated protein kinase (AMPK) activity to improve mitochondrial fatty acid β-oxidation (FAO) by regulating acetyl-CoA carboxylase (ACC) and carnitine palmitoyltransferase 1A (CPT1A)-mediated FAO pathway. In addition, P2Y2R deficiency increased peroxisome proliferator-activated gamma co-activator-1α (PGC-1α)-mediated mitochondrial biogenesis. Conclusively, P2Y2R deficiency ameliorated HFD-induced hepatic steatosis by enhancing FAO through AMPK signaling and PGC-1α pathway, suggesting P2Y2R as a promising therapeutic target for NAFLD.
Collapse
Affiliation(s)
- Theodomir Dusabimana
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 52727, Korea; (T.D.); (E.J.P.); (J.J.); (K.J.); (S.P.Y.); (H.J.K.)
- Department of Convergence Medical Sciences, Institute of Health Sciences, Gyeongsang National University Graduate School, Jinju 52727, Korea
| | - Eun Jung Park
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 52727, Korea; (T.D.); (E.J.P.); (J.J.); (K.J.); (S.P.Y.); (H.J.K.)
| | - Jihyun Je
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 52727, Korea; (T.D.); (E.J.P.); (J.J.); (K.J.); (S.P.Y.); (H.J.K.)
| | - Kyuho Jeong
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 52727, Korea; (T.D.); (E.J.P.); (J.J.); (K.J.); (S.P.Y.); (H.J.K.)
| | - Seung Pil Yun
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 52727, Korea; (T.D.); (E.J.P.); (J.J.); (K.J.); (S.P.Y.); (H.J.K.)
- Department of Convergence Medical Sciences, Institute of Health Sciences, Gyeongsang National University Graduate School, Jinju 52727, Korea
| | - Hye Jung Kim
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 52727, Korea; (T.D.); (E.J.P.); (J.J.); (K.J.); (S.P.Y.); (H.J.K.)
- Department of Convergence Medical Sciences, Institute of Health Sciences, Gyeongsang National University Graduate School, Jinju 52727, Korea
| | - Hwajin Kim
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 52727, Korea; (T.D.); (E.J.P.); (J.J.); (K.J.); (S.P.Y.); (H.J.K.)
- Correspondence: (H.K.); (S.W.P.); Tel.: +82-55-772-8070 (H.K.); +82-55-772-8073 (S.W.P.)
| | - Sang Won Park
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 52727, Korea; (T.D.); (E.J.P.); (J.J.); (K.J.); (S.P.Y.); (H.J.K.)
- Department of Convergence Medical Sciences, Institute of Health Sciences, Gyeongsang National University Graduate School, Jinju 52727, Korea
- Correspondence: (H.K.); (S.W.P.); Tel.: +82-55-772-8070 (H.K.); +82-55-772-8073 (S.W.P.)
| |
Collapse
|
50
|
Kasper P, Breuer S, Hoffmann T, Vohlen C, Janoschek R, Schmitz L, Appel S, Fink G, Hünseler C, Quaas A, Demir M, Lang S, Steffen HM, Martin A, Schramm C, Bürger M, Mahabir E, Goeser T, Dötsch J, Hucklenbruch-Rother E, Bae-Gartz I. Maternal Exercise Mediates Hepatic Metabolic Programming via Activation of AMPK-PGC1α Axis in the Offspring of Obese Mothers. Cells 2021; 10:1247. [PMID: 34069390 PMCID: PMC8158724 DOI: 10.3390/cells10051247] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/11/2021] [Accepted: 05/11/2021] [Indexed: 12/13/2022] Open
Abstract
Maternal obesity is associated with an increased risk of hepatic metabolic dysfunction for both mother and offspring and targeted interventions to address this growing metabolic disease burden are urgently needed. This study investigates whether maternal exercise (ME) could reverse the detrimental effects of hepatic metabolic dysfunction in obese dams and their offspring while focusing on the AMP-activated protein kinase (AMPK), representing a key regulator of hepatic metabolism. In a mouse model of maternal western-style-diet (WSD)-induced obesity, we established an exercise intervention of voluntary wheel-running before and during pregnancy and analyzed its effects on hepatic energy metabolism during developmental organ programming. ME prevented WSD-induced hepatic steatosis in obese dams by alterations of key hepatic metabolic processes, including activation of hepatic ß-oxidation and inhibition of lipogenesis following increased AMPK and peroxisome-proliferator-activated-receptor-γ-coactivator-1α (PGC-1α)-signaling. Offspring of exercised dams exhibited a comparable hepatic metabolic signature to their mothers with increased AMPK-PGC1α-activity and beneficial changes in hepatic lipid metabolism and were protected from WSD-induced adipose tissue accumulation and hepatic steatosis in later life. In conclusion, this study demonstrates that ME provides a promising strategy to improve the metabolic health of both obese mothers and their offspring and highlights AMPK as a potential metabolic target for therapeutic interventions.
Collapse
Affiliation(s)
- Philipp Kasper
- Clinic for Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (P.K.); (S.L.); (H.-M.S.); (A.M.); (C.S.); (M.B.); (T.G.)
| | - Saida Breuer
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Thorben Hoffmann
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Christina Vohlen
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Ruth Janoschek
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Lisa Schmitz
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Sarah Appel
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Gregor Fink
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Christoph Hünseler
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Alexander Quaas
- Department of Pathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany;
| | - Münevver Demir
- Charité Campus Mitte and Campus Virchow Clinic, Department of Hepatology and Gastroenterology, Charité University Medicine Berlin, D-13353 Berlin, Germany;
| | - Sonja Lang
- Clinic for Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (P.K.); (S.L.); (H.-M.S.); (A.M.); (C.S.); (M.B.); (T.G.)
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Hans-Michael Steffen
- Clinic for Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (P.K.); (S.L.); (H.-M.S.); (A.M.); (C.S.); (M.B.); (T.G.)
| | - Anna Martin
- Clinic for Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (P.K.); (S.L.); (H.-M.S.); (A.M.); (C.S.); (M.B.); (T.G.)
| | - Christoph Schramm
- Clinic for Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (P.K.); (S.L.); (H.-M.S.); (A.M.); (C.S.); (M.B.); (T.G.)
| | - Martin Bürger
- Clinic for Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (P.K.); (S.L.); (H.-M.S.); (A.M.); (C.S.); (M.B.); (T.G.)
| | - Esther Mahabir
- Comparative Medicine, Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, D-50937 Cologne, Germany;
| | - Tobias Goeser
- Clinic for Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (P.K.); (S.L.); (H.-M.S.); (A.M.); (C.S.); (M.B.); (T.G.)
| | - Jörg Dötsch
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Eva Hucklenbruch-Rother
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Inga Bae-Gartz
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| |
Collapse
|