1
|
Barbier-Torres L, Luque-Urbano M, Chhimwal J, Robinson AE, Fernández-Ramos D, Lopitz-Otsoa F, Van Eyk JE, Millet O, Mato JM, Lu SC. Fructose-induced progression of steatohepatitis involves disrupting aldolase B-AMPK signaling in methionine adenosyltransferase 1A deficient mice. Metabolism 2025; 165:156154. [PMID: 39922455 DOI: 10.1016/j.metabol.2025.156154] [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: 10/25/2024] [Revised: 01/23/2025] [Accepted: 02/03/2025] [Indexed: 02/10/2025]
Abstract
OBJECTIVE Aldolases (ALDO) are sensors that regulate AMPK via binding to fructose 1,6-biphosphate (FBP), an intermediate of glucose and fructose metabolism. Fructose consumption is linked to metabolic dysfunction-associated steatotic liver disease (MASLD) progression but whether ALDO-AMPK signaling is involved is unknown. Methionine adenosyltransferase alpha 1 (Mat1a) knockout (KO) mice have low hepatic S-adenosylmethionine (SAMe) level and spontaneously develop steatohepatitis. ALDOB methylation has not been reported and here we investigated whether SAMe level regulates ALDOB and ALDOB-AMPK signaling and whether fructose feeding accelerates MASLD progression by disrupting ALDOB-AMPK signaling. METHODS Mass spectrometry identified ALDOB methylation sites and recombinant in vitro approaches assessed how methylation at those sites affects ALDOB oligomerization and activity. Primary hepatocytes cultured with high/low glucose and/or fructose and wild type (WT) and Mat1a KO mice fed with a high-fructose diet examined AMPK-ALDOB signaling and MASLD progression. RESULTS In Mat1a KO livers ALDOB R173 is hypomethylated while ALDOB activity is enhanced. Recombinant ALDOB is methylated at R173 and R304 by protein arginine methyltransferase 1. Low hepatic SAMe level results in hypomethylated ALDOB, which favors the tetrameric form that has higher enzymatic activity, and higher capacity to signal to activate AMPK. Fructose, independently of glucose levels, inhibited AMPK activity and induced lipid accumulation in hepatocytes. Mat1a KO mice have hyperactivated AMPK and fructose feeding inhibits it, enhancing the accumulation of fat in the liver and the progression of MASLD. CONCLUSION Hepatic SAMe levels regulate ALDOB oligomeric state and enzymatic activity impacting on AMPK signaling and fructose-induced MASLD progression.
Collapse
Affiliation(s)
- Lucía Barbier-Torres
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - María Luque-Urbano
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - Jyoti Chhimwal
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Aaron E Robinson
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - David Fernández-Ramos
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - Fernando Lopitz-Otsoa
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - Jennifer E Van Eyk
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Oscar Millet
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - José M Mato
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain.
| | - Shelly C Lu
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| |
Collapse
|
2
|
Kim S, Han HJ, Rho H, Kang S, Mukherjee S, Kim J, Kim D, Ko HW, Lim SM, Im SS, Chung JY, Song J. Ebastine-mediated destabilization of E3 ligase MKRN1 protects against metabolic dysfunction-associated steatohepatitis. Cell Mol Life Sci 2025; 82:66. [PMID: 39888429 PMCID: PMC11785899 DOI: 10.1007/s00018-024-05535-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 11/08/2024] [Accepted: 11/29/2024] [Indexed: 02/01/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a chronic condition encompassing metabolic dysfunction-associated steatotic liver (MASL) and metabolic dysfunction-associated steatohepatitis (MASH), which can progress to fibrosis, cirrhosis, or hepatocellular carcinoma (HCC). The heterogeneous and complex nature of MASLD complicates optimal drug development. Ebastine, an antihistamine, exhibits antitumor activity in various types of cancer. However, its effects on MASH remain unexplored. In the present study, we identified ebastine as a potential treatment for MASH. Our results indicated that ebastine acts as a novel MKRN1 inhibitor by promoting MKRN1 destabilization through self-ubiquitination, leading to AMP-activated protein kinase (AMPK) activation. Ebastine appeared to bind to the C-terminal domain of MKRN1, particularly at residues R298 and K360. Notably, Mkrn1 knockout (KO) mice demonstrated resistance to MASH, including obesity, steatosis, inflammation, and fibrosis under high-fat-high-fructose diet (HFHFD) conditions. Additionally, liver-specific Mkrn1 knockdown using AAV8 alleviated MASH symptoms in HFHFD-fed mice, implicating MKRN1 as a potential therapeutic target. Consistent with these findings, treatment with ebastine significantly reduced the risk of MASH in HFHFD-fed mice, with a decrease in MKRN1 expression and an increase in AMPK activity. Our study suggests that ebastine binds to MKRN1, promoting its destabilization and subsequent degradation by stimulating its ubiquitination. This enhances AMPK stability and activity, suppressing lipid accumulation, inflammation, and fibrosis. Moreover, the knockout of Mkrn1 mice decreased the risk of MASH, suggesting that ebastine could be a promising therapeutic agent for the treatment of MASH.
Collapse
Affiliation(s)
- Seungyeon Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hyun-Ji Han
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hyunjin Rho
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Subin Kang
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Sulagna Mukherjee
- Department of Physiology, Keimyung University School of Medicine, Daegu, 42601, Republic of Korea
| | - Jiwoo Kim
- Therapeutics & Biotechnology Division, Drug Discovery Platform Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Republic of Korea
| | - Doyoun Kim
- Therapeutics & Biotechnology Division, Drug Discovery Platform Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Republic of Korea
- Medicinal Chemistry and Pharmacology, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, 07292, Republic of Korea
| | - Hyuk Wan Ko
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
- Department of Biochemistry, College of Life Science and Technology, Institute for Bio-medical Convergence Science and Technology, Yonsei University, Seoul, Republic of Korea
| | - Sang Min Lim
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, 07292, Republic of Korea
| | - Seung-Soon Im
- Department of Physiology, Keimyung University School of Medicine, Daegu, 42601, Republic of Korea
| | - Joon-Yong Chung
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jaewhan Song
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea.
- Department of Biochemistry, College of Life Science and Technology, Institute for Bio-medical Convergence Science and Technology, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
3
|
Zakaria SS, Hanafy SM. Unraveling the Beneficial Role of Resveratrol in Fructose-Induced Non-Alcoholic Steatohepatitis with a Focus on the AMPK/Nrf2 Signaling Axis. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:139. [PMID: 39859121 PMCID: PMC11767180 DOI: 10.3390/medicina61010139] [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: 12/19/2024] [Revised: 01/03/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
Background and Objectives: High fructose intake is associated with non-alcoholic fatty liver disease (NAFLD), a chronic liver disease that is on the rise worldwide. New alternatives for treatment, such as bioactive phytochemicals, are needed. The aim of this study was to investigate the beneficial role of resveratrol in treating non-alcoholic steatohepatitis (NASH). Materials and Methods: Sixty male albino rats were allocated to three groups: group I, the normal control group; group II, the fructose-enriched diet group (FED), which was fed a 70% fructose diet for six weeks to induce NASH; and group III, the resveratrol-FED group (RES + FED), which was given the same FED diet plus an oral dose of 70 mg/kg resveratrol (RES) every day for an additional six weeks. We performed histological evaluations and assessed blood lipids and liver enzymes to study resveratrol's impact on NASH. Quantitative real-time PCR was used to assess the mRNA expression of nuclear factor E2-related factor 2 (Nrf2) in the liver samples. ELISA was used to measure Beclin 1, AMPK, IL-6, and the DNA-binding activity of Nrf2. Oxidative stress indicators, including GSH, SOD, and MDA, were evaluated spectrophotometrically. Results: Resveratrol effectively alleviated the biochemical and histopathological abnormalities associated with NASH, improving autophagy by raising Beclin 1 levels while reducing inflammation by decreasing IL-6 levels. Furthermore, resveratrol restored the liver architecture and the oxidative balance, as evidenced by the decreased MDA levels and improved antioxidant status via elevated GSH and SOD activities, as well as the activation of the AMPK/Nrf2 signaling axis. Conclusions: This study specifically examines resveratrol's therapeutic effects in a high-fructose diet-induced NASH model, focusing on the AMPK/Nrf2 signaling pathway to address oxidative stress and autophagy, providing novel insights into its molecular mechanism of action. Resveratrol reduces NASH by boosting autophagy and activating the AMPK/Nrf2 pathway. These findings underscore the potential of resveratrol as a promising therapeutic agent that can support treatment alongside conventional medications in the management of non-alcoholic steatohepatitis (NASH).
Collapse
Affiliation(s)
- Soha S. Zakaria
- Department of Biochemistry, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 13317, Saudi Arabia
| | - Safaa M. Hanafy
- Department of Anatomy and Physiology, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 13317, Saudi Arabia;
| |
Collapse
|
4
|
Milošević N, Milanović M, Medić Stojanoska M, Tipmanee V, Smyrnioudis I, Dedoussis GV, Milić N. Triterpenoids from Chios Mastiha Resin Against MASLD-A Molecular Docking Survey. Curr Issues Mol Biol 2025; 47:51. [PMID: 39852166 PMCID: PMC11763944 DOI: 10.3390/cimb47010051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/08/2025] [Accepted: 01/11/2025] [Indexed: 01/26/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease without an approved pharmacological approach for its prevention/treatment. Based on the modified Delphi process, NAFLD was redefined as metabolic dysfunction-associated steatotic liver disease (MASLD) to highlight the metabolic aspect of liver pathogenesis. Chios mastiha (Pistacia lentiscus var. Chia, Anacardiaceae) resin demonstrated promising results in MASLD treatment. In this paper, molecular docking was applied to test 16 compounds from Chios mastiha as potential ligands for the receptors GR, LXRα, LXRβ, PPARα PPARγ, MC4R, AMPK, and VEGFR2, whose up- and down-regulation interfere with MASLD development and progression. The observed compounds had moderate and high affinity for LXR, GR, MC4R, and PPARγ in comparison to proven ligands, while their affinity for PPARα, AMPK, and VEGFR was less pronounced. The combination of active compounds from Chios mastiha rather than a single molecule may have a superior ability to control the intertwined MASLD metabolic pathways.
Collapse
Affiliation(s)
- Nataša Milošević
- Department of Pharmacy, Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia; (N.M.); (N.M.)
| | - Maja Milanović
- Department of Pharmacy, Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia; (N.M.); (N.M.)
| | - Milica Medić Stojanoska
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia;
- University Clinical Center of Vojvodina, 21000 Novi Sad, Serbia
| | - Varomyalin Tipmanee
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand;
| | | | - George V. Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University of Athens, 17671 Athens, Greece;
| | - Nataša Milić
- Department of Pharmacy, Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia; (N.M.); (N.M.)
| |
Collapse
|
5
|
Malicka A, Ali A, MacCannell ADV, Roberts LD. Brown and beige adipose tissue-derived metabokine and lipokine inter-organ signalling in health and disease. Exp Physiol 2024. [PMID: 39591977 DOI: 10.1113/ep092008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024]
Abstract
Adipose tissue has an established endocrine function through the secretion of adipokines. However, a role for bioactive metabolites and lipids, termed metabokines and lipokines, is emerging in adipose tissue-mediated autocrine, paracrine and endocrine signalling and inter-organ communication. Traditionally seen as passive entities, metabolites are now recognized for their active roles in regulating cellular signalling and local and systemic metabolism. Distinct from white adipose tissue, specific endocrine functions have been attributed to thermogenic brown and beige adipose tissues. Brown and beige adipose tissues have been identified as sources of metabokines and lipokines, which influence diverse metabolic pathways, such as fatty acid β-oxidation, mitochondrial function and glucose homeostasis, across a range of tissues, including skeletal muscle, adipose tissue and heart. This review explores the intricate signalling mechanisms of brown and beige adipose tissue-derived metabokines and lipokines, emphasizing their roles in maintaining metabolic homeostasis and their potential dysregulation in metabolic diseases. Furthermore, we discuss the therapeutic potential of targeting these pathways, proposing that precise modulation of metabokine receptors and transporters could offer superior specificity and efficacy in comparison to conventional approaches, such as β-adrenergic signalling-stimulated activation of brown adipose tissue thermogenesis. Understanding the complex interactions between adipokines, metabokines and lipokines is essential for developing a systems-level approach to new interventions for metabolic disorders, underscoring the need for continued research in this rapidly evolving field.
Collapse
Affiliation(s)
- Anna Malicka
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Aysha Ali
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Amanda D V MacCannell
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Lee D Roberts
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| |
Collapse
|
6
|
Pollard AE. New concepts in the roles of AMPK in adipocyte stem cell biology. Essays Biochem 2024; 68:349-361. [PMID: 39175418 DOI: 10.1042/ebc20240008] [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: 05/31/2024] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 08/24/2024]
Abstract
Obesity is a major risk factor for many life-threatening diseases. Adipose tissue dysfunction is emerging as a driving factor in the transition from excess adiposity to comorbidities such as metabolic-associated fatty liver disease, cardiovascular disease, Type 2 diabetes and cancer. However, the transition from healthy adipose expansion to the development of these conditions is poorly understood. Adipose stem cells, residing in the vasculature and stromal regions of subcutaneous and visceral depots, are responsible for the expansion and maintenance of organ function, and are now recognised as key mediators of pathological transformation. Impaired tissue expansion drives inflammation, dysregulation of endocrine function and the deposition of lipids in the liver, muscle and around vital organs, where it is toxic. Contrary to previous hypotheses, it is the promotion of healthy adipose tissue expansion and function, not inhibition of adipogenesis, that presents the most attractive therapeutic strategy in the treatment of metabolic disease. AMP-activated protein kinase, a master regulator of energy homeostasis, has been regarded as one such target, due to its central role in adipose tissue lipid metabolism, and its apparent inhibition of adipogenesis. However, recent studies utilising AMP-activated protein kinase (AMPK)-specific compounds highlight a more subtle, time-dependent role for AMPK in the process of adipogenesis, and in a previously unexplored repression of leptin, independent of adipocyte maturity. In this article, I discuss historic evidence for AMPK-mediated adipogenesis inhibition and the multi-faceted roles for AMPK in adipose tissue.
Collapse
Affiliation(s)
- Alice E Pollard
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, U.K
| |
Collapse
|
7
|
Gadiraju B, Magisetty J, Kondreddy V. Transcription factor ETV4 plays a critical role in the development of non-alcoholic fatty liver disease. Int J Biol Macromol 2024; 282:137235. [PMID: 39500423 DOI: 10.1016/j.ijbiomac.2024.137235] [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: 08/09/2024] [Revised: 10/25/2024] [Accepted: 11/01/2024] [Indexed: 11/12/2024]
Abstract
The Angiopoietin-like 4 (ANGPTL4) and ETS Variant Transcription Factor 4 (ETV4) are involved in the metabolic transition and carcinogenesis in the liver. However, the role of ETV4 in the development of non-alcoholic fatty liver disease (NAFLD) is currently unknown. Our study reveals that ETV4 expression was upregulated in the diet-induced non-alcoholic fatty liver disease, and plays a critical role in the dysregulated lipid metabolism. We demonstrate a mechanism by which ANGPTL4 regulates lipid homeostasis via involving the AMPK/ETV4 axis. Transient knockdown of ETV4 abolished the ANGPTL4-induced expression of Srebp1c, Acc and Fasn. Insulin treatment potentially increased the physical association of ETV4 with SREBP1, and promotes nuclear translocation and transcriptional activity of SREBP1. In addition, we show that combined therapy with omega-3 fatty acids and diacylglycerol O-acyltransferase inhibitor 1 (DGAT1) inhibitor (A-922500) counteracted the ANGPTL4-ETV4 axis-induced lipogenesis in vitro, and in vivo in obese mice via activation of GPR120-βarrestin2-AMPK pathway. Finally, we demonstrate that targeted pharmacologic therapy using GalNac-ETV4 siRNA that specifically inhibits ETV4 gene expression in the liver protects against diet-induced NAFLD, obesity and dyslipidemia. Hence, our study reveal previously unrecognized role of ETV4 in the NAFLD, and provides rationale targeting ETV4 to treat NAFLD.
Collapse
Affiliation(s)
- Bhavani Gadiraju
- Department of Biochemistry, Central University of Punjab, Bathinda, India
| | - Jhansi Magisetty
- Department of Zoology, Central University of Punjab, Bathinda., India.
| | - Vijay Kondreddy
- Department of Biochemistry, Central University of Punjab, Bathinda, India.
| |
Collapse
|
8
|
Song GY, Kim SM, Back S, Yang SB, Yang YM. Atractylodes Lancea and Its Constituent, Atractylodin, Ameliorates Metabolic Dysfunction-Associated Steatotic Liver Disease via AMPK Activation. Biomol Ther (Seoul) 2024; 32:778-792. [PMID: 39391981 PMCID: PMC11535289 DOI: 10.4062/biomolther.2024.083] [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: 05/22/2024] [Revised: 07/14/2024] [Accepted: 07/24/2024] [Indexed: 10/12/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), which encompasses a spectrum of conditions ranging from simple steatosis to hepatocellular carcinoma, is a growing global health concern associated with insulin resistance. Since there are limited treatment options for MASLD, this study investigated the therapeutic potential of Atractylodes lancea, a traditional herbal remedy for digestive disorders in East Asia, and its principal component, atractylodin, in treating MASLD. Following 8 weeks of high-fat diet (HFD) feeding, mice received oral doses of 30, 60, or 120 mg/kg of Atractylodes lancea. In HFD-fed mice, Atractylodes lancea treatment reduced the body weight; serum triglyceride, total cholesterol, and alanine aminotransferase levels; and hepatic lipid content. Furthermore, Atractylodes lancea significantly ameliorated fasting serum glucose, fasting serum insulin, and homeostatic model assessment of insulin resistance levels in response to HFD. Additionally, a glucose tolerance test demonstrated improved glucose homeostasis. Treatment with 5 or 10 mg/kg atractylodin also resulted in anti-obesity, anti-steatosis, and glucose-lowering effects. Atractylodin treatment resulted in the downregulation of key lipogenic genes (Srebf1, Fasn, Scd2, and Dgat2) and the upregulation of genes regulated by peroxisome proliferator-activated receptor-α. Notably, the molecular docking model suggested a robust binding affinity between atractylodin and AMP-activated protein kinase (AMPK). Atractylodin activated AMPK, which contributed to SREBP1c regulation. In conclusion, our results revealed that Atractylodes lancea and atractylodin activated the AMPK signaling pathway, leading to improvements in HFD-induced obesity, fatty liver, and glucose intolerance. This study suggests that the phytochemical, atractylodin, can be a treatment option for MASLD.
Collapse
Affiliation(s)
- Ga Yeon Song
- Department of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Sun Myoung Kim
- Department of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea
- KNU Innovative Drug Development Research Team for Intractable Diseases (BK21 Four), Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Seungil Back
- Department of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Seung-Bo Yang
- Department of Korean Internal Medicine, College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea
| | - Yoon Mee Yang
- Department of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea
- KNU Innovative Drug Development Research Team for Intractable Diseases (BK21 Four), Kangwon National University, Chuncheon 24341, Republic of Korea
| |
Collapse
|
9
|
Ashraf N, Van Nostrand JL. Fine-tuning AMPK in physiology and disease using point-mutant mouse models. Dis Model Mech 2024; 17:dmm050798. [PMID: 39136185 PMCID: PMC11340815 DOI: 10.1242/dmm.050798] [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] [Indexed: 08/17/2024] Open
Abstract
AMP-activated protein kinase (AMPK) is an evolutionarily conserved serine/threonine kinase that monitors the cellular energy status to adapt it to the fluctuating nutritional and environmental conditions in an organism. AMPK plays an integral part in a wide array of physiological processes, such as cell growth, autophagy and mitochondrial function, and is implicated in diverse diseases, including cancer, metabolic disorders, cardiovascular diseases and neurodegenerative diseases. AMPK orchestrates many different physiological outcomes by phosphorylating a broad range of downstream substrates. However, the importance of AMPK-mediated regulation of these substrates in vivo remains an ongoing area of investigation to better understand its precise role in cellular and metabolic homeostasis. Here, we provide a comprehensive overview of our understanding of the kinase function of AMPK in vivo, as uncovered from mouse models that harbor phosphorylation mutations in AMPK substrates. We discuss some of the inherent limitations of these mouse models, highlight the broader implications of these studies for understanding human health and disease, and explore the valuable insights gained that could inform future therapeutic strategies for the treatment of metabolic and non-metabolic disorders.
Collapse
Affiliation(s)
- Naghmana Ashraf
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jeanine L. Van Nostrand
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
10
|
McGlone ER, Hope DCD, Davies I, Dore M, Goldin R, Jones B, Liu Z, Li JV, Vorkas PA, Khoo B, Carling D, Minnion J, Bloom SR, Tan TMM. Chronic treatment with glucagon-like peptide-1 and glucagon receptor co-agonist causes weight loss-independent improvements in hepatic steatosis in mice with diet-induced obesity. Biomed Pharmacother 2024; 176:116888. [PMID: 38861859 DOI: 10.1016/j.biopha.2024.116888] [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: 05/03/2024] [Revised: 05/28/2024] [Accepted: 06/03/2024] [Indexed: 06/13/2024] Open
Abstract
OBJECTIVES Co-agonists at the glucagon-like peptide-1 and glucagon receptors (GLP1R/GCGR) show promise as treatments for metabolic dysfunction-associated steatotic liver disease (MASLD). Although most co-agonists to date have been heavily GLP1R-biased, glucagon directly acts on the liver to reduce fat content. The aims of this study were to investigate a GCGR-biased co-agonist as treatment for hepatic steatosis in mice. METHODS Mice with diet-induced obesity (DIO) were treated with Dicretin, a GLP1/GCGR co-agonist with high potency at the GCGR, Semaglutide (GLP1R monoagonist) or food restriction over 24 days, such that their weight loss was matched. Hepatic steatosis, glucose tolerance, hepatic transcriptomics, metabolomics and lipidomics at the end of the study were compared with Vehicle-treated mice. RESULTS Dicretin lead to superior reduction of hepatic lipid content when compared to Semaglutide or equivalent weight loss by calorie restriction. Markers of glucose tolerance and insulin resistance improved in all treatment groups. Hepatic transcriptomic and metabolomic profiling demonstrated many changes that were unique to Dicretin-treated mice. These include some known targets of glucagon signaling and others with as yet unclear physiological significance. CONCLUSIONS Our study supports the development of GCGR-biased GLP1/GCGR co-agonists for treatment of MASLD and related conditions.
Collapse
Affiliation(s)
- Emma Rose McGlone
- Department of Surgery and Cancer, Imperial College London, London, UK; Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - David C D Hope
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Iona Davies
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Marian Dore
- Genomics facility, MRC Laboratory of Medical Sciences (LMS), Imperial College London, London, UK
| | - Rob Goldin
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Ben Jones
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Zhigang Liu
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Jia V Li
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Panagiotis A Vorkas
- Institute of Applied Biosciences, Centre for Research and Technology Hellas (INAB|CERTH), Thessaloniki 57001, Greece; School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, London, UK
| | - Bernard Khoo
- Endocrinology, Division of Medicine, University College London, London, UK
| | - David Carling
- Cellular Stress group, MRC LMS, Imperial College London, London, UK
| | - James Minnion
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Stephen R Bloom
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Tricia M-M Tan
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| |
Collapse
|
11
|
Nunes JRC, O'Dwyer C, Ghorbani P, Smith TKT, Chauhan S, Robert-Gostlin V, Girouard MD, Viollet B, Foretz M, Fullerton MD. Myeloid AMPK signaling restricts fibrosis but is not required for metformin improvements during CDAHFD-induced NASH in mice. J Lipid Res 2024; 65:100564. [PMID: 38762124 PMCID: PMC11222943 DOI: 10.1016/j.jlr.2024.100564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 05/07/2024] [Accepted: 05/11/2024] [Indexed: 05/20/2024] Open
Abstract
Metabolic programming underpins inflammation and liver macrophage activation in the setting of chronic liver disease. Here, we sought to identify the role of an important metabolic regulator, AMP-activated protein kinase (AMPK), specifically within myeloid cells during the progression of non-alcoholic steatohepatitis (NASH) and whether treatment with metformin, a firstline therapy for diabetes and activator of AMPK could stem disease progression. Male and female Prkaa1fl/fl/Prkaa2fl/fl (Flox) control and Flox-LysM-Cre+ (MacKO) mice were fed a low-fat control or a choline-deficient, amino acid defined 45% Kcal high-fat diet (CDAHFD) for 8 weeks, where metformin was introduced in the drinking water (50 or 250 mg/kg/day) for the last 4 weeks. Hepatic steatosis and fibrosis were dramatically increased in response to CDAHFD-feeding compared to low-fat control. While myeloid AMPK signaling had no effect on markers of hepatic steatosis or circulating markers, fibrosis as measured by total liver collagen was significantly elevated in livers from MacKO mice, independent of sex. Although treatment with 50 mg/kg/day metformin had no effect on any parameter, intervention with 250 mg/kg/day metformin completely ameliorated hepatic steatosis and fibrosis in both male and female mice. While the protective effect of metformin was associated with lower final body weight, and decreased expression of lipogenic and Col1a1 transcripts, it was independent of myeloid AMPK signaling. These results suggest that endogenous AMPK signaling in myeloid cells, both liver-resident and infiltrating, acts to restrict fibrogenesis during CDAHFD-induced NASH progression but is not the mechanism by which metformin improves markers of NASH.
Collapse
Affiliation(s)
- Julia R C Nunes
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, Centre for Infection, Immunity and Inflammation, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
| | - Conor O'Dwyer
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, Centre for Infection, Immunity and Inflammation, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
| | - Peyman Ghorbani
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, Centre for Infection, Immunity and Inflammation, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
| | - Tyler K T Smith
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, Centre for Infection, Immunity and Inflammation, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
| | - Samarth Chauhan
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, Centre for Infection, Immunity and Inflammation, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
| | - Victoria Robert-Gostlin
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, Centre for Infection, Immunity and Inflammation, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
| | - Madison D Girouard
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, Centre for Infection, Immunity and Inflammation, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
| | - Benoit Viollet
- Université Paris cité, CNRS, Inserm, Institut Cochin, Paris, France
| | - Marc Foretz
- Université Paris cité, CNRS, Inserm, Institut Cochin, Paris, France
| | - Morgan D Fullerton
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, Centre for Infection, Immunity and Inflammation, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada; Centre for Catalysis Research and Innovation, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
12
|
Bustraan S, Bennett J, Whilding C, Pennycook BR, Smith D, Barr AR, Read J, Carling D, Pollard A. AMP-activated protein kinase activation suppresses leptin expression independently of adipogenesis in primary murine adipocytes. Biochem J 2024; 481:345-362. [PMID: 38314646 PMCID: PMC11088909 DOI: 10.1042/bcj20240003] [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: 01/06/2024] [Revised: 02/01/2024] [Accepted: 02/04/2024] [Indexed: 02/06/2024]
Abstract
Adipogenesis, defined as the development of mature adipocytes from stem cell precursors, is vital for the expansion, turnover and health of adipose tissue. Loss of adipogenic potential in adipose stem cells, or impairment of adipogenesis is now recognised as an underlying cause of adipose tissue dysfunction and is associated with metabolic disease. In this study, we sought to determine the role of AMP-activated protein kinase (AMPK), an evolutionarily conserved master regulator of energy homeostasis, in adipogenesis. Primary murine adipose-derived stem cells were treated with a small molecule AMPK activator (BI-9774) during key phases of adipogenesis, to determine the effect of AMPK activation on adipocyte commitment, maturation and function. To determine the contribution of the repression of lipogenesis by AMPK in these processes, we compared the effect of pharmacological inhibition of acetyl-CoA carboxylase (ACC). We show that AMPK activation inhibits adipogenesis in a time- and concentration-dependent manner. Transient AMPK activation during adipogenic commitment leads to a significant, ACC-independent, repression of adipogenic transcription factor expression. Furthermore, we identify a striking, previously unexplored inhibition of leptin gene expression in response to both short-term and chronic AMPK activation irrespective of adipogenesis. These findings reveal that in addition to its effect on adipogenesis, AMPK activation switches off leptin gene expression in primary mouse adipocytes independently of adipogenesis. Our results identify leptin expression as a novel target of AMPK through mechanisms yet to be identified.
Collapse
Affiliation(s)
- Sophia Bustraan
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, U.K
- Medical Research Council Laboratory of Medical Sciences, London, U.K
| | - Jane Bennett
- Medical Research Council Laboratory of Medical Sciences, London, U.K
| | - Chad Whilding
- Medical Research Council Laboratory of Medical Sciences, London, U.K
| | | | - David Smith
- Emerging Innovations Unit, Discovery Sciences, R&D, AstraZeneca, Cambridge, U.K
| | - Alexis R. Barr
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, U.K
- Medical Research Council Laboratory of Medical Sciences, London, U.K
| | - Jon Read
- Mechanistic and Structural Biology, Biopharmaceuticals R&D, AstraZeneca, Cambridge, U.K
| | - David Carling
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, U.K
- Medical Research Council Laboratory of Medical Sciences, London, U.K
| | - Alice Pollard
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, U.K
| |
Collapse
|
13
|
Yaseen U, Hwang S, Park S, Kim SB, Lee HJ, Cha JY. New Insights into the Role of KLF10 in Tissue Fibrosis. Int J Mol Sci 2024; 25:1276. [PMID: 38279278 PMCID: PMC10816924 DOI: 10.3390/ijms25021276] [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: 12/14/2023] [Revised: 01/14/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024] Open
Abstract
Fibrosis, characterized by excessive extracellular matrix accumulation, disrupts normal tissue architecture, causes organ dysfunction, and contributes to numerous chronic diseases. This review focuses on Krüppel-like factor 10 (KLF10), a transcription factor significantly induced by transforming growth factor-β (TGF-β), and its role in fibrosis pathogenesis and progression across various tissues. KLF10, initially identified as TGF-β-inducible early gene-1 (TIEG1), is involved in key biological processes including cell proliferation, differentiation, apoptosis, and immune responses. Our analysis investigated KLF10 gene and protein structures, interaction partners, and context-dependent functions in fibrotic diseases. This review highlights recent findings that underscore KLF10 interaction with pivotal signaling pathways, such as TGF-β, and the modulation of gene expression in fibrotic tissues. We examined the dual role of KLF10 in promoting and inhibiting fibrosis depending on tissue type and fibrotic context. This review also discusses the therapeutic potential of targeting KLF10 in fibrotic diseases, based on its regulatory role in key pathogenic mechanisms. By consolidating current research, this review aims to enhance the understanding of the multifaceted role of KLF10 in fibrosis and stimulate further research into its potential as a therapeutic target in combating fibrotic diseases.
Collapse
Affiliation(s)
- Uzma Yaseen
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea; (U.Y.); (S.P.); (S.-B.K.)
| | - Soonjae Hwang
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, College of Medicine, Gachon University, Incheon 21999, Republic of Korea;
| | - Sangbin Park
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea; (U.Y.); (S.P.); (S.-B.K.)
| | - Soo-Bin Kim
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea; (U.Y.); (S.P.); (S.-B.K.)
| | - Ho-Jae Lee
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea; (U.Y.); (S.P.); (S.-B.K.)
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, College of Medicine, Gachon University, Incheon 21999, Republic of Korea;
| | - Ji-Young Cha
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea; (U.Y.); (S.P.); (S.-B.K.)
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, College of Medicine, Gachon University, Incheon 21999, Republic of Korea;
| |
Collapse
|
14
|
Hughey CC, Bracy DP, Rome FI, Goelzer M, Donahue EP, Viollet B, Foretz M, Wasserman DH. Exercise training adaptations in liver glycogen and glycerolipids require hepatic AMP-activated protein kinase in mice. Am J Physiol Endocrinol Metab 2024; 326:E14-E28. [PMID: 37938177 PMCID: PMC11193517 DOI: 10.1152/ajpendo.00289.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/06/2023] [Accepted: 11/06/2023] [Indexed: 11/09/2023]
Abstract
Regular exercise elicits adaptations in glucose and lipid metabolism that allow the body to meet energy demands of subsequent exercise bouts more effectively and mitigate metabolic diseases including fatty liver. Energy discharged during the acute exercise bouts that comprise exercise training may be a catalyst for liver adaptations. During acute exercise, liver glycogenolysis and gluconeogenesis are accelerated to supply glucose to working muscle. Lower liver energy state imposed by gluconeogenesis and related pathways activates AMP-activated protein kinase (AMPK), which conserves ATP partly by promoting lipid oxidation. This study tested the hypothesis that AMPK is necessary for liver glucose and lipid adaptations to training. Liver-specific AMPKα1α2 knockout (AMPKα1α2fl/fl+AlbCre) mice and littermate controls (AMPKα1α2fl/fl) completed sedentary and exercise training protocols. Liver nutrient fluxes were quantified at rest or during acute exercise following training. Liver metabolites and molecular regulators of metabolism were assessed. Training increased liver glycogen in AMPKα1α2fl/fl mice, but not in AMPKα1α2fl/fl+AlbCre mice. The inability to increase glycogen led to lower glycogenolysis, glucose production, and circulating glucose during acute exercise in trained AMPKα1α2fl/fl+AlbCre mice. Deletion of AMPKα1α2 attenuated training-induced declines in liver diacylglycerides. In particular, training lowered the concentration of unsaturated and elongated fatty acids comprising diacylglycerides in AMPKα1α2fl/fl mice, but not in AMPKα1α2fl/fl+AlbCre mice. Training increased liver triacylglycerides and the desaturation and elongation of fatty acids in triacylglycerides of AMPKα1α2fl/fl+AlbCre mice. These lipid responses were independent of differences in tricarboxylic acid cycle fluxes. In conclusion, AMPK is required for liver training adaptations that are critical to glucose and lipid metabolism.NEW & NOTEWORTHY This study shows that the energy sensor and transducer, AMP-activated protein kinase (AMPK), is necessary for an exercise training-induced: 1) increase in liver glycogen that is necessary for accelerated glycogenolysis during exercise, 2) decrease in liver glycerolipids independent of tricarboxylic acid (TCA) cycle flux, and 3) decline in the desaturation and elongation of fatty acids comprising liver diacylglycerides. The mechanisms defined in these studies have implications for use of regular exercise or AMPK-activators in patients with fatty liver.
Collapse
Affiliation(s)
- Curtis C Hughey
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, United States
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Deanna P Bracy
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
- Mouse Metabolic Phenotyping Center, Vanderbilt University, Nashville, Tennessee, United States
| | - Ferrol I Rome
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, United States
| | - Mickael Goelzer
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - E Patrick Donahue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Benoit Viollet
- Université Paris Cité, CNRS, Inserm, Institut Cochin, Paris, France
| | - Marc Foretz
- Université Paris Cité, CNRS, Inserm, Institut Cochin, Paris, France
| | - David H Wasserman
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
- Mouse Metabolic Phenotyping Center, Vanderbilt University, Nashville, Tennessee, United States
| |
Collapse
|
15
|
Li J, Wang X, Shi L, Liu B, Sheng Z, Chang S, Cai X, Shan G. A Mammalian Conserved Circular RNA CircLARP1B Regulates Hepatocellular Carcinoma Metastasis and Lipid Metabolism. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305902. [PMID: 37953462 PMCID: PMC10787103 DOI: 10.1002/advs.202305902] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/14/2023] [Indexed: 11/14/2023]
Abstract
Circular RNAs (circRNAs) have emerged as crucial regulators in physiology and human diseases. However, evolutionarily conserved circRNAs with potent functions in cancers are rarely reported. In this study, a mammalian conserved circRNA circLARP1B is identified to play critical roles in hepatocellular carcinoma (HCC). Patients with high circLARP1B levels have advanced prognostic stage and poor overall survival. CircLARP1B facilitates cellular metastatic properties and lipid accumulation through promoting fatty acid synthesis in HCC. CircLARP1B deficient mice exhibit reduced metastasis and less lipid accumulation in an induced HCC model. Multiple lines of evidence demonstrate that circLARP1B binds to heterogeneous nuclear ribonucleoprotein D (HNRNPD) in the cytoplasm, and thus affects the binding of HNRNPD to sensitive transcripts including liver kinase B1 (LKB1) mRNA. This regulation causes decreased LKB1 mRNA stability and lower LKB1 protein levels. Antisense oligodeoxynucleotide complementary to theHNRNPD binding sites in circLARP1B increases the HNRNPD binding to LKB1 mRNA. Through the HNRNPD-LKB1-AMPK pathway, circLARP1B promotes HCC metastasis and lipid accumulation. Results from AAV8-mediated hepatocyte-directed knockdown of circLARP1B or Lkb1 in mouse models also demonstrate critical roles of hepatocytic circLARP1B regulatory pathway in HCC metastasis and lipid accumulation, and indicate that circLARP1B may be potential target of HCC treatment.
Collapse
Affiliation(s)
- Jingxin Li
- Department of Laboratory MedicineThe First Affiliated Hospital of USTCThe CAS Key Laboratory of Innate Immunity and Chronic DiseaseSchool of Basic Medical SciencesDivision of Life Science and MedicineUniversity of Science and Technology of ChinaHefei230027China
| | - Xiaolin Wang
- Department of Laboratory MedicineThe First Affiliated Hospital of USTCThe CAS Key Laboratory of Innate Immunity and Chronic DiseaseSchool of Basic Medical SciencesDivision of Life Science and MedicineUniversity of Science and Technology of ChinaHefei230027China
| | - Liang Shi
- Department of General SurgerySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhou310016China
| | - Boqiang Liu
- Department of General SurgerySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhou310016China
| | - Zhiyong Sheng
- School of Life ScienceBengbu Medical CollegeBengbu233030China
| | - Shuhui Chang
- Department of Laboratory MedicineThe First Affiliated Hospital of USTCThe CAS Key Laboratory of Innate Immunity and Chronic DiseaseSchool of Basic Medical SciencesDivision of Life Science and MedicineUniversity of Science and Technology of ChinaHefei230027China
| | - Xiujun Cai
- Department of General SurgerySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhou310016China
| | - Ge Shan
- Department of Laboratory MedicineThe First Affiliated Hospital of USTCThe CAS Key Laboratory of Innate Immunity and Chronic DiseaseSchool of Basic Medical SciencesDivision of Life Science and MedicineUniversity of Science and Technology of ChinaHefei230027China
- Department of Pulmonary and Critical Care MedicineRegional Medical Center for National Institute of Respiratory DiseasesSir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhou310016China
| |
Collapse
|
16
|
Promsuwan S, Sawamoto K, Xu L, Nagashimada M, Nagata N, Takiyama Y. A natural Nrf2 activator glucoraphanin improves hepatic steatosis in high-fat diet-induced obese male mice associated with AMPK activation. Diabetol Int 2024; 15:86-98. [PMID: 38264234 PMCID: PMC10800329 DOI: 10.1007/s13340-023-00658-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 08/09/2023] [Indexed: 01/25/2024]
Abstract
Genetic and pharmacological activation of the transcription factor nuclear factor, erythroid derived 2, like 2 (Nrf2) alleviates high-fat diet (HFD)-induced obesity in mice; however, synthetic Nrf2 activators are not clinically available due to safety concerns. Dietary glucoraphanin (GR), a naturally occurring compound found in cruciferous vegetables that activates Nrf2 and induces its target antioxidant genes. We previously demonstrated that GR increased thermogenesis and mitigated HFD-induced obesity in lean healthy mice. In this study, we investigated the therapeutic effects of GR on pre-existing obesity and associated metabolic disorders, such as hepatic steatosis, with or without low-fat dietary intervention. Eight-week-old male C57BL/6J mice were fed an HFD for 9 weeks to induce obesity. Subsequently, these obese mice were fed either the HFD or a normal chow diet, supplemented with or without GR, for an additional 11 weeks. GR supplementation did not decrease the body weight of HFD-fed mice; however, it significantly reduced plasma alanine aminotransferase and aspartate aminotransferase levels and hepatic triglyceride accumulation. These improvements in liver damage by GR were associated with decreased expression levels of fatty acid synthesis genes and proinflammatory chemokine genes, suppressed c-Jun N-terminal kinase activation, and reduced proinflammatory phenotypes of macrophages in the liver. Moreover, metabolome analysis identified increased hepatic levels of adenosine 5'-monophosphate (AMP) in HFD-GR mice compared with those in HFD mice, which agreed with increased phosphorylation levels of AMP-activated protein kinase. Our results show that GR may have a therapeutic potential for treating obesity-associated hepatic steatosis. Supplementary Information The online version contains supplementary material available at 10.1007/s13340-023-00658-6.
Collapse
Affiliation(s)
- Suratsawadee Promsuwan
- Division of Diabetes, Department of Internal Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka Higashi, Asahikawa, 078-8510 Japan
| | - Kazuki Sawamoto
- Division of Diabetes, Department of Internal Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka Higashi, Asahikawa, 078-8510 Japan
| | - Liang Xu
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035 Zhejiang China
| | - Mayumi Nagashimada
- Division of Health Sciences, Graduate School of Medical Science, Kanazawa University, Kanazawa, 920-0942 Japan
| | - Naoto Nagata
- Department of Cellular and Molecular Function Analysis, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, 920-8640 Japan
| | - Yumi Takiyama
- Division of Diabetes, Department of Internal Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka Higashi, Asahikawa, 078-8510 Japan
| |
Collapse
|
17
|
Lee HS, Jung JI, Hwang JS, Hwang MO, Kim EJ. Cydonia oblonga Miller fruit extract exerts an anti-obesity effect in 3T3-L1 adipocytes by activating the AMPK signaling pathway. Nutr Res Pract 2023; 17:1043-1055. [PMID: 38053822 PMCID: PMC10694414 DOI: 10.4162/nrp.2023.17.6.1043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/27/2023] [Accepted: 07/13/2023] [Indexed: 12/07/2023] Open
Abstract
BACKGROUND/OBJECTIVES The fruit of Cydonia oblonga Miller (COM) is used traditionally in Mediterranean region medicine to prevent or treat obesity, but its mechanism of action is still unclear. Beyond a demonstrated anti-obesity effect, the fruit was tested for the mechanism of adipogenesis in 3T3-L1 preadipocytes. MATERIALS/METHODS 3T3-L1 preadipocytes were cultured for 8 days with COM fruit extract (COME) at different concentrations (0-600 µg/mL) with adipocyte differentiation medium. The cell viability was measured using an MTT assay; triglyceride (TG) was stained with Oil Red O. The expression levels of the adipogenesis-related genes and protein expression were analyzed by reverse transcription polymerase chain reaction and Western blotting, respectively. RESULTS COME inhibited intracellular TG accumulation during adipogenesis. A COME treatment in 3T3-L1 cells induced upregulation of the adenosine monophosphate-activated protein kinase (AMPK)α phosphorylation and downregulation of the adipogenic transcription factors, such as sterol regulatory element-binding protein 1c, peroxisome proliferator-activated receptor γ, and CCAAT/enhancer binding protein α. The COME treatment reduced the mRNA expression of fatty acyl synthetase, adenosine triphosphate-citrate lyase, adipocyte protein 2, and lipoprotein lipase. It increased the mRNA expression of hormone-sensitive lipase and carnitine palmitoyltransferase I in 3T3-L1 cells. CONCLUSIONS COME inhibits adipogenesis via the AMPK signaling pathways. COME may be used to prevent and treat obesity.
Collapse
Affiliation(s)
- Hyun Sook Lee
- Department of Food Science & Nutrition, Dongseo University, Busan 47011, Korea
| | - Jae In Jung
- Industry coupled Cooperation Center for Bio Healthcare Materials, Hallym University, Chuncheon 24252, Korea
| | | | | | - Eun Ji Kim
- Industry coupled Cooperation Center for Bio Healthcare Materials, Hallym University, Chuncheon 24252, Korea
| |
Collapse
|
18
|
Tutunchi H, Ebrahimi-Mameghani M, Hosseinzadeh-Attar MJ, Roshanravan N, Mobasseri M, Najafipour F, Naeini F, Naghshi S, Asghari S, Akbarzadeh M, Soleimanzadeh H, Ostadrahimi A. Effects of oleoylethanolamide supplementation on the expression of lipid metabolism-related genes and serum NRG4 levels in patients with non-alcoholic fatty liver disease: A randomized controlled trial. Clin Nutr ESPEN 2023; 58:311-319. [PMID: 38057021 DOI: 10.1016/j.clnesp.2023.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 08/08/2023] [Accepted: 10/16/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND This study investigated the effects of oleoylethanolamide (OEA) supplementation on the expression levels of SIRT1, AMPK, PGC-1α, PPAR-γ, CEBP-α and CEBP-β genes and serum neuregulin 4 (NRG4) levels in patients with non-alcoholic fatty liver diseases (NAFLD). METHODS Sixty obese patients with NAFLD were equally allocated into either OEA or placebo group for 12 weeks. The mRNA expression levels of genes were determined using the reverse transcription polymerase chain reaction (RT-PCR) technique. Serum NRG4 level was also assessed using an enzyme-linked immunosorbent assay (ELISA) kit. RESULTS At the endpoint, mRNA expression levels of SIRT1(p = 0.001), PGC-1α (p = 0.011) and AMPK (p = 0.019) were significantly higher in the OEA group compared to placebo group. However, no significant differences were observed in the expression levels of PPAR-γ, CEBP-α and CEBP-β between the two groups. Serum NRG4 levels significantly increased in the OEA group compared with the placebo group after controlling for confounders (p = 0.027). In the OEA group, significant relationships were found between percent of changes in the expression levels of the SIRT1, AMPK and PGC-1α as well as serum NRG4 level with percent of changes in some anthropometric measures. Moreover, in the intervention group, percent of changes in high-density lipoprotein cholesterol was positively correlated with percent of changes in the expression levels of the SIRT1 and AMPK. While, percent of changes in triglyceride was inversely correlated with percent of changes in the expression levels of SIRT1. CONCLUSION OEA could beneficially affect expression levels of some lipid metabolism-related genes and serum NRG4 level. "REGISTERED UNDER IRANIAN REGISTRY OF CLINICAL TRIALS IDENTIFIER NO: IRCT20090609002017N32".
Collapse
Affiliation(s)
- Helda Tutunchi
- Endocrine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mehrangiz Ebrahimi-Mameghani
- Nutrition Research Center, Department of Biochemistry and Diet Therapy, Faculty of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammad Javad Hosseinzadeh-Attar
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran.
| | - Neda Roshanravan
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Majid Mobasseri
- Endocrine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Farzad Najafipour
- Endocrine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Fatemeh Naeini
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran.
| | - Sina Naghshi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Samira Asghari
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Moloud Akbarzadeh
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Hamid Soleimanzadeh
- Department of Applied Chemistry, Faculty of Chemistry, University of Tabriz, Tabriz, Iran.
| | - Alireza Ostadrahimi
- Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
19
|
Oh E, Lee J, Cho S, Kim SW, Won K, Shin WS, Gwak SH, Ha J, Jeon SY, Park JH, Song IS, Thoudam T, Lee IK, Kim S, Choi SY, Kim KT. Gossypetin Prevents the Progression of Nonalcoholic Steatohepatitis by Regulating Oxidative Stress and AMP-Activated Protein Kinase. Mol Pharmacol 2023; 104:214-229. [PMID: 37595967 DOI: 10.1124/molpharm.123.000675] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 07/22/2023] [Accepted: 07/25/2023] [Indexed: 08/20/2023] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is a severe liver metabolic disorder, however, there are still no effective and safe drugs for its treatment. Previous clinical trials used various therapeutic approaches to target individual pathologic mechanisms, but these approaches were unsuccessful because of the complex pathologic causes of NASH. Combinatory therapy in which two or more drugs are administered simultaneously to patients with NASH, however, carries the risk of side effects associated with each individual drug. To solve this problem, we identified gossypetin as an effective dual-targeting agent that activates AMP-activated protein kinase (AMPK) and decreases oxidative stress. Administration of gossypetin decreased hepatic steatosis, lobular inflammation and liver fibrosis in the liver tissue of mice with choline-deficient high-fat diet and methionine-choline deficient diet (MCD) diet-induced NASH. Gossypetin functioned directly as an antioxidant agent, decreasing hydrogen peroxide and palmitate-induced oxidative stress in the AML12 cells and liver tissue of MCD diet-fed mice without regulating the antioxidant response factors. In addition, gossypetin acted as a novel AMPK activator by binding to the allosteric drug and metabolite site, which stabilizes the activated structure of AMPK. Our findings demonstrate that gossypetin has the potential to serve as a novel therapeutic agent for nonalcoholic fatty liver disease /NASH. SIGNIFICANCE STATEMENT: This study demonstrates that gossypetin has preventive effect to progression of nonalcoholic steatohepatitis (NASH) as a novel AMP-activated protein kinase (AMPK) activator and antioxidants. Our findings indicate that simultaneous activation of AMPK and oxidative stress using gossypetin has the potential to serve as a novel therapeutic approach for nonalcoholic fatty liver disease /NASH patients.
Collapse
Affiliation(s)
- Eunji Oh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Jae Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Sungji Cho
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Sung Wook Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Kyung Won
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Won Sik Shin
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Seung Hee Gwak
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Joohun Ha
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - So Yeon Jeon
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Jin-Hyang Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Im-Sook Song
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Themis Thoudam
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - In-Kyu Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Seonyong Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Se-Young Choi
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Kyong-Tai Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| |
Collapse
|
20
|
Kim J, Lee SK, Jeong SY, You H, Han SD, Park S, Kim S, Kim TM. Multifaceted action of stem cell-derived extracellular vesicles for nonalcoholic steatohepatitis. J Control Release 2023; 364:S0168-3659(23)00706-X. [PMID: 39491172 DOI: 10.1016/j.jconrel.2023.10.045] [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: 06/22/2023] [Revised: 10/24/2023] [Accepted: 10/27/2023] [Indexed: 11/05/2024]
Abstract
Nonalcoholic steatohepatitis (NASH) is a chronic liver disease associated with metabolic syndrome. Extracellular vesicles (EVs) are essential signaling mediators containing functional biomolecules. EVs are secreted from various cell types, and recent studies have shown that mesenchymal stem cell-derived EVs have therapeutic potential against immune and metabolic diseases. In this study, we investigated whether EVs from induced mesenchymal stem cells (iMSC-EVs) regulate AMPK signaling and lipid metabolism using cell-based studies and two different mouse models of NASH (methionine/choline-deficient diet-induced and ob/ob mice). Protein analysis revealed that iMSC-EVs carry cargo proteins with the potential to regulate lipid metabolism. iMSC-EVs inhibited free fatty acid release from adipose tissues by downregulating the activity of lipolytic genes in NASH. In addition, iMSC-EVs improved hepatic steatosis by modulating AMPK signaling, which plays essential role in metabolic homeostasis in the liver. Moreover, iMSC-EVs reduced CD36 expression, contributing to the blockade of free fatty acid transport to the liver of NASH mice. Finally, iMSC-EVs reduced inflammation, endoplasmic reticulum stress, and apoptosis while promoting hepatic regeneration of the NASH liver. In conclusion, iMSC-EVs can potentially serve as cell-free therapeutics for NASH owing to their multifaceted modality.
Collapse
Affiliation(s)
- Jimin Kim
- Brexogen Research Center, Brexogen Inc., Songpa-Gu, Seoul 05855, South Korea
| | - Seul Ki Lee
- Brexogen Research Center, Brexogen Inc., Songpa-Gu, Seoul 05855, South Korea
| | - Seon-Yeong Jeong
- Brexogen Research Center, Brexogen Inc., Songpa-Gu, Seoul 05855, South Korea
| | - Haedeun You
- Brexogen Research Center, Brexogen Inc., Songpa-Gu, Seoul 05855, South Korea
| | - Sang-Deok Han
- Brexogen Research Center, Brexogen Inc., Songpa-Gu, Seoul 05855, South Korea
| | - Somi Park
- Brexogen Research Center, Brexogen Inc., Songpa-Gu, Seoul 05855, South Korea
| | - Soo Kim
- Brexogen Research Center, Brexogen Inc., Songpa-Gu, Seoul 05855, South Korea
| | - Tae Min Kim
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang, Gangwon-do 25354, South Korea; Institutes of Green-Bio Science and Technology, Seoul National University, Pyeongchang, Gangwon-do 25354, South Korea.
| |
Collapse
|
21
|
McGlone ER, Siebert M, Dore M, Hope DCD, Davies I, Owen B, Khoo B, Goldin R, Carling D, Bloom S, Le Gall M, Tan TM. Sleeve gastrectomy causes weight-loss independent improvements in hepatic steatosis. Liver Int 2023; 43:1890-1900. [PMID: 37208943 PMCID: PMC10947097 DOI: 10.1111/liv.15614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 03/15/2023] [Accepted: 05/05/2023] [Indexed: 05/21/2023]
Abstract
BACKGROUND AND AIMS Sleeve gastrectomy (VSG) leads to improvement in hepatic steatosis, associated with weight loss. The aims of this study were to investigate whether VSG leads to weight-loss independent improvements in liver steatosis in mice with diet-induced obesity (DIO); and to metabolically and transcriptomically profile hepatic changes in mice undergoing VSG. METHODS Mice with DIO were treated with VSG, sham surgery with subsequent food restriction to weight-match to the VSG group (Sham-WM), or sham surgery with return to unrestricted diet (Sham-Ad lib). Hepatic steatosis, glucose tolerance, insulin and glucagon resistance, and hepatic transcriptomics were investigated at the end of the study period and treatment groups were compared with mice undergoing sham surgery only (Sham-Ad lib). RESULTS VSG led to much greater improvement in liver steatosis than Sham-WM (liver triglyceride mg/mg 2.5 ± 0.1, 2.1 ± 0.2, 1.6 ± 0.1 for Sham-AL, Sham-WM and VSG respectively; p = 0.003). Homeostatic model assessment of insulin resistance was improved following VSG only (51.2 ± 8.8, 36.3 ± 5.3, 22.3 ± 6.1 for Sham-AL, Sham-WM and VSG respectively; p = 0.03). The glucagon-alanine index, a measure of glucagon resistance, fell with VSG but was significantly increased in Sham-WM (9.8 ± 1.7, 25.8 ± 4.6 and 5.2 ± 1.2 in Sham Ad-lib, Sham-WM and VSG respectively; p = 0.0003). Genes downstream of glucagon receptor signalling which govern fatty acid synthesis (Acaca, Acacb, Me1, Acly, Fasn and Elovl6) were downregulated following VSG but upregulated in Sham-WM. CONCLUSIONS Changes in glucagon sensitivity may contribute to weight-loss independent improvements in hepatic steatosis following VSG.
Collapse
Affiliation(s)
- Emma Rose McGlone
- Department of Surgery and CancerImperial College LondonLondonUK
- Department of Metabolism, Digestion and ReproductionImperial College LondonLondonUK
| | - Matthieu Siebert
- Centre de Recherche sur l'Inflammation, UMRS1149, Inserm, Université Paris CitéParisFrance
| | - Marian Dore
- Genomics FacilityMRC London Institute of Medical Sciences (LMS), Imperial College LondonLondonUK
| | - David C. D. Hope
- Department of Metabolism, Digestion and ReproductionImperial College LondonLondonUK
| | - Iona Davies
- Department of Metabolism, Digestion and ReproductionImperial College LondonLondonUK
| | - Bryn Owen
- Department of Metabolism, Digestion and ReproductionImperial College LondonLondonUK
| | - Bernard Khoo
- Division of MedicineUniversity College London, Royal Free HospitalLondonUK
| | - Rob Goldin
- Department of Metabolism, Digestion and ReproductionImperial College LondonLondonUK
| | - Dave Carling
- Cellular Stress GroupMRC LMS, Imperial College LondonLondonUK
| | - Stephen Bloom
- Department of Metabolism, Digestion and ReproductionImperial College LondonLondonUK
| | - Maude Le Gall
- Centre de Recherche sur l'Inflammation, UMRS1149, Inserm, Université Paris CitéParisFrance
| | - Tricia M‐M. Tan
- Department of Metabolism, Digestion and ReproductionImperial College LondonLondonUK
| |
Collapse
|
22
|
Sun H, Kemper JK. MicroRNA regulation of AMPK in nonalcoholic fatty liver disease. Exp Mol Med 2023; 55:1974-1981. [PMID: 37653034 PMCID: PMC10545736 DOI: 10.1038/s12276-023-01072-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/10/2023] [Accepted: 06/13/2023] [Indexed: 09/02/2023] Open
Abstract
Obesity-associated nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease and is the leading cause of liver failure and death. The function of AMP-activated protein kinase (AMPK), a master energy sensor, is aberrantly reduced in NAFLD, but the underlying mechanisms are not fully understood. Increasing evidence indicates that aberrantly expressed microRNAs (miRs) are associated with impaired AMPK function in obesity and NAFLD. In this review, we discuss the emerging evidence that miRs have a role in reducing AMPK activity in NAFLD and nonalcoholic steatohepatitis (NASH), a severe form of NAFLD. We also discuss the underlying mechanisms of the aberrant expression of miRs that can negatively impact AMPK, as well as the therapeutic potential of targeting the miR-AMPK pathway for NAFLD/NASH.
Collapse
Affiliation(s)
- Hao Sun
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Jongsook Kim Kemper
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
23
|
Astre G, Atlan T, Goshtchevsky U, Oron-Gottesman A, Smirnov M, Shapira K, Velan A, Deelen J, Levy T, Levanon EY, Harel I. Genetic perturbation of AMP biosynthesis extends lifespan and restores metabolic health in a naturally short-lived vertebrate. Dev Cell 2023; 58:1350-1364.e10. [PMID: 37321215 DOI: 10.1016/j.devcel.2023.05.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 03/09/2023] [Accepted: 05/23/2023] [Indexed: 06/17/2023]
Abstract
During aging, the loss of metabolic homeostasis drives a myriad of pathologies. A central regulator of cellular energy, the AMP-activated protein kinase (AMPK), orchestrates organismal metabolism. However, direct genetic manipulations of the AMPK complex in mice have, so far, produced detrimental phenotypes. Here, as an alternative approach, we alter energy homeostasis by manipulating the upstream nucleotide pool. Using the turquoise killifish, we mutate APRT, a key enzyme in AMP biosynthesis, and extend the lifespan of heterozygous males. Next, we apply an integrated omics approach to show that metabolic functions are rejuvenated in old mutants, which also display a fasting-like metabolic profile and resistance to high-fat diet. At the cellular level, heterozygous cells exhibit enhanced nutrient sensitivity, reduced ATP levels, and AMPK activation. Finally, lifelong intermittent fasting abolishes the longevity benefits. Our findings suggest that perturbing AMP biosynthesis may modulate vertebrate lifespan and propose APRT as a promising target for promoting metabolic health.
Collapse
Affiliation(s)
- Gwendoline Astre
- Department of Genetics, the Silberman Institute, the Hebrew University of Jerusalem, Givat Ram, Jerusalem 91904, Israel
| | - Tehila Atlan
- Department of Genetics, the Silberman Institute, the Hebrew University of Jerusalem, Givat Ram, Jerusalem 91904, Israel
| | - Uri Goshtchevsky
- Department of Genetics, the Silberman Institute, the Hebrew University of Jerusalem, Givat Ram, Jerusalem 91904, Israel
| | - Adi Oron-Gottesman
- Department of Genetics, the Silberman Institute, the Hebrew University of Jerusalem, Givat Ram, Jerusalem 91904, Israel
| | - Margarita Smirnov
- Central Fish Health Laboratory, Department of Fisheries and Aquaculture, Ministry of Agriculture and Rural Development, Nir David 10803, Israel
| | - Kobi Shapira
- Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel
| | - Ariel Velan
- Department of Genetics, the Silberman Institute, the Hebrew University of Jerusalem, Givat Ram, Jerusalem 91904, Israel
| | - Joris Deelen
- Max Planck Institute for Biology of Ageing, Cologne 50931, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), University of Cologne, Cologne 50931, Germany
| | - Tomer Levy
- Department of Genetics, the Silberman Institute, the Hebrew University of Jerusalem, Givat Ram, Jerusalem 91904, Israel
| | - Erez Y Levanon
- Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel
| | - Itamar Harel
- Department of Genetics, the Silberman Institute, the Hebrew University of Jerusalem, Givat Ram, Jerusalem 91904, Israel.
| |
Collapse
|
24
|
Likitnukul S, Tepaarmorndech S, Kaewamatawong T, Yangchum A, Duangtha C, Jongjang P, Mangmool S, Pinthong D, Isaka M. Pyridylnidulin exerts anti-diabetic properties and improves non-alcoholic fatty liver disease in diet-induced obesity mice. Front Mol Biosci 2023; 10:1208215. [PMID: 37426418 PMCID: PMC10324605 DOI: 10.3389/fmolb.2023.1208215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/15/2023] [Indexed: 07/11/2023] Open
Abstract
Introduction: Non-alcoholic fatty liver disease (NAFLD) is one of the metabolic disorders related to the pathophysiology of type 2 diabetes mellitus (T2DM). Therapeutic strategies are focused on the improvement of energy balance and lifestyle modification. Additionally, the derivative of the bioactive fungal metabolite is of interest to provide health benefits, especially in obese and pre-diabetic conditions. In our screening of anti-diabetic compounds from fungal metabolites and semisynthetic derivatives, a depsidone derivative, namely pyridylnidulin (PN), showed potent glucose uptake-inducing activity. The present study aimed to investigate the liver lipid metabolism and anti-diabetic properties of PN in diet-induced obesity mice. Methods: Male C57BL/6 mice were induced obesity and pre-diabetic conditions by dietary intervention with a high-fat diet (HFD) for 6 weeks. These obese mice were orally administered with PN (40 or 120 mg/kg), metformin (150 mg/kg), or vehicle for 4 weeks. Glucose tolerance, plasma adipocytokines, hepatic gene and protein expressions were assessed after treatment. Results: Improved glucose tolerance and reduced fasting blood glucose levels were found in the PN and metformin-treated mice. Additionally, hepatic triglyceride levels were consistent with the histopathological steatosis score regarding hepatocellular hypertrophy in the PN and metformin groups. The levels of plasma adipocytokines such as tumor necrosis factor-α (TNF-α) and monocyte chemoattractant protein-1 (MCP-1) were reduced in the PN (120 mg/kg) and metformin-treated mice. In addition, hepatic gene expression involved in lipid metabolism, including lipogenic enzymes was significantly reduced in the PN (120 mg/kg) and metformin-treated mice. The increased protein expression levels of phosphorylated AMP-activated protein kinase (p-AMPK) was also found in PN and metformin-treated mice. Discussion: Considering the increased p-AMPK protein expression levels in PN and metformin-treated mice were revealed as the underlying mechanisms to improve metabolic parameters. These results suggested that PN provided the health benefit to slow the progression of NAFLD and T2DM in obese and pre-diabetic conditions.
Collapse
Affiliation(s)
- Sutharinee Likitnukul
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | | | - Theerayuth Kaewamatawong
- Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Arunrat Yangchum
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani, Thailand
| | - Chanathip Duangtha
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Pimrapat Jongjang
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Supachoke Mangmool
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Darawan Pinthong
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Masahiko Isaka
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani, Thailand
| |
Collapse
|
25
|
Chen Y, Wang W, Morgan MP, Robson T, Annett S. Obesity, non-alcoholic fatty liver disease and hepatocellular carcinoma: current status and therapeutic targets. Front Endocrinol (Lausanne) 2023; 14:1148934. [PMID: 37361533 PMCID: PMC10286797 DOI: 10.3389/fendo.2023.1148934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 05/16/2023] [Indexed: 06/28/2023] Open
Abstract
Obesity is a global epidemic and overwhelming evidence indicates that it is a risk factor for numerous cancers, including hepatocellular carcinoma (HCC), the third leading cause of cancer-related deaths worldwide. Obesity-associated hepatic tumorigenesis develops from nonalcoholic fatty liver disease (NAFLD), progressing to nonalcoholic steatohepatitis (NASH), cirrhosis and ultimately to HCC. The rising incidence of obesity is resulting in an increased prevalence of NAFLD and NASH, and subsequently HCC. Obesity represents an increasingly important underlying etiology of HCC, in particular as the other leading causes of HCC such as hepatitis infection, are declining due to effective treatments and vaccines. In this review, we provide a comprehensive overview of the molecular mechanisms and cellular signaling pathways involved in the pathogenesis of obesity-associated HCC. We summarize the preclinical experimental animal models available to study the features of NAFLD/NASH/HCC, and the non-invasive methods to diagnose NAFLD, NASH and early-stage HCC. Finally, since HCC is an aggressive tumor with a 5-year survival of less than 20%, we will also discuss novel therapeutic targets for obesity-associated HCC and ongoing clinical trials.
Collapse
Affiliation(s)
- Yinshuang Chen
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland
- College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Weipeng Wang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Maria P. Morgan
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland
| | - Tracy Robson
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland
| | - Stephanie Annett
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland
| |
Collapse
|
26
|
Penfold L, Woods A, Pollard AE, Arizanova J, Pascual-Navarro E, Muckett PJ, Dore MH, Montoya A, Whilding C, Fets L, Mokochinski J, Constantin TA, Varela-Carver A, Leach DA, Bevan CL, Nikitin AY, Hall Z, Carling D. AMPK activation protects against prostate cancer by inducing a catabolic cellular state. Cell Rep 2023; 42:112396. [PMID: 37061917 PMCID: PMC10576838 DOI: 10.1016/j.celrep.2023.112396] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 12/02/2022] [Accepted: 03/30/2023] [Indexed: 04/17/2023] Open
Abstract
Emerging evidence indicates that metabolic dysregulation drives prostate cancer (PCa) progression and metastasis. AMP-activated protein kinase (AMPK) is a master regulator of metabolism, although its role in PCa remains unclear. Here, we show that genetic and pharmacological activation of AMPK provides a protective effect on PCa progression in vivo. We show that AMPK activation induces PGC1α expression, leading to catabolic metabolic reprogramming of PCa cells. This catabolic state is characterized by increased mitochondrial gene expression, increased fatty acid oxidation, decreased lipogenic potential, decreased cell proliferation, and decreased cell invasiveness. Together, these changes inhibit PCa disease progression. Additionally, we identify a gene network involved in cell cycle regulation that is inhibited by AMPK activation. Strikingly, we show a correlation between this gene network and PGC1α gene expression in human PCa. Taken together, our findings support the use of AMPK activators for clinical treatment of PCa to improve patient outcome.
Collapse
Affiliation(s)
- Lucy Penfold
- MRC London Institute of Medical Sciences, Hammersmith Hospital Campus, Imperial College London, London W12 0NN, UK.
| | - Angela Woods
- MRC London Institute of Medical Sciences, Hammersmith Hospital Campus, Imperial College London, London W12 0NN, UK
| | - Alice E Pollard
- Institute of Clinical Sciences, Imperial College London, London, UK
| | - Julia Arizanova
- MRC London Institute of Medical Sciences, Hammersmith Hospital Campus, Imperial College London, London W12 0NN, UK
| | - Eneko Pascual-Navarro
- MRC London Institute of Medical Sciences, Hammersmith Hospital Campus, Imperial College London, London W12 0NN, UK
| | - Phillip J Muckett
- MRC London Institute of Medical Sciences, Hammersmith Hospital Campus, Imperial College London, London W12 0NN, UK
| | - Marian H Dore
- MRC London Institute of Medical Sciences, Hammersmith Hospital Campus, Imperial College London, London W12 0NN, UK
| | - Alex Montoya
- MRC London Institute of Medical Sciences, Hammersmith Hospital Campus, Imperial College London, London W12 0NN, UK
| | - Chad Whilding
- MRC London Institute of Medical Sciences, Hammersmith Hospital Campus, Imperial College London, London W12 0NN, UK
| | - Louise Fets
- MRC London Institute of Medical Sciences, Hammersmith Hospital Campus, Imperial College London, London W12 0NN, UK
| | - Joao Mokochinski
- MRC London Institute of Medical Sciences, Hammersmith Hospital Campus, Imperial College London, London W12 0NN, UK
| | - Theodora A Constantin
- Imperial Centre for Translational and Experimental Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Anabel Varela-Carver
- Imperial Centre for Translational and Experimental Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Damien A Leach
- Imperial Centre for Translational and Experimental Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Charlotte L Bevan
- Imperial Centre for Translational and Experimental Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Alexander Yu Nikitin
- Department of Biomedical Sciences and Cornell Stem Cell Program, Cornell University, Ithaca, NY, USA
| | - Zoe Hall
- Biomolecular Medicine, Division of Systems Medicine, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, UK
| | - David Carling
- MRC London Institute of Medical Sciences, Hammersmith Hospital Campus, Imperial College London, London W12 0NN, UK; Institute of Clinical Sciences, Imperial College London, London, UK.
| |
Collapse
|
27
|
Au-Yeung KKW, Shang Y, Wijerathne CUB, Madduma Hewage S, Siow YL, O K. Acute Kidney Injury Induces Oxidative Stress and Hepatic Lipid Accumulation through AMPK Signaling Pathway. Antioxidants (Basel) 2023; 12:antiox12040883. [PMID: 37107258 PMCID: PMC10135179 DOI: 10.3390/antiox12040883] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/27/2023] [Accepted: 04/02/2023] [Indexed: 04/08/2023] Open
Abstract
Acute kidney injury (AKI) often impairs the function of other organs leading to distant organ injury. The liver is the major organ that regulates metabolism and lipid homeostasis in the body. It has been reported that AKI causes liver injury with increased oxidative stress, inflammatory response and steatosis. In the present study, we investigated the mechanisms by which ischemia-reperfusion-induced AKI caused hepatic lipid accumulation. Kidney ischemia (45 min)-reperfusion (24 h) led to a significant increase in plasma creatinine and transaminase in Sprague Dawley rats, indicating kidney and liver injury. Histological and biochemical analyses revealed hepatic lipid accumulation with a significant elevation of triglyceride and cholesterol levels in the liver. This was accompanied by a decreased AMP-activated protein kinase (AMPK) phosphorylation, indicating the reduced activation of AMPK, which is an energy sensor that regulates lipid metabolism. The expression of AMPK-regulated genes that were responsible for fatty acid oxidation (CPTIα, ACOX) was significantly decreased, while the expression of lipogenesis genes (SREPB-1c, ACC1) was significantly elevated. The oxidative stress biomarker malondialdehyde was elevated in the plasma and liver. Incubation of HepG2 cells with an oxidative stress inducer hydrogen peroxide inhibited AMPK phosphorylation and caused cellular lipid accumulation. This was accompanied by decreased expression of genes responsible for fatty acid oxidation and increased expression of genes responsible for lipogenesis. These results suggest that AKI elicits hepatic lipid accumulation through decreased fatty acid metabolism and increased lipogenesis. Oxidative stress may contribute, in part, to the downregulation of the AMPK signaling pathway leading to hepatic lipid accumulation and injury.
Collapse
Affiliation(s)
- Kathy K. W. Au-Yeung
- St. Boniface Hospital Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Animal Science, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Yue Shang
- St. Boniface Hospital Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Animal Science, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Charith U. B. Wijerathne
- St. Boniface Hospital Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Animal Science, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Susara Madduma Hewage
- St. Boniface Hospital Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Physiology & Pathophysiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Yaw L. Siow
- Department of Physiology & Pathophysiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- Agriculture and Agri-Food Canada, St. Boniface Hospital Research Centre, Winnipeg, MB R2H 2A6, Canada
| | - Karmin O
- St. Boniface Hospital Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Animal Science, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Department of Physiology & Pathophysiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
28
|
Steinberg GR, Hardie DG. New insights into activation and function of the AMPK. Nat Rev Mol Cell Biol 2023; 24:255-272. [PMID: 36316383 DOI: 10.1038/s41580-022-00547-x] [Citation(s) in RCA: 311] [Impact Index Per Article: 155.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/23/2022] [Indexed: 11/06/2022]
Abstract
The classical role of AMP-activated protein kinase (AMPK) is as a cellular energy sensor activated by falling energy status, signalled by increases in AMP to ATP and ADP to ATP ratios. Once activated, AMPK acts to restore energy homeostasis by promoting ATP-producing catabolic pathways while inhibiting energy-consuming processes. In this Review, we provide an update on this canonical (AMP/ADP-dependent) activation mechanism, but focus mainly on recently described non-canonical pathways, including those by which AMPK senses the availability of glucose, glycogen or fatty acids and by which it senses damage to lysosomes and nuclear DNA. We also discuss new findings on the regulation of carbohydrate and lipid metabolism, mitochondrial and lysosomal homeostasis, and DNA repair. Finally, we discuss the role of AMPK in cancer, obesity, diabetes, nonalcoholic steatohepatitis (NASH) and other disorders where therapeutic targeting may exert beneficial effects.
Collapse
Affiliation(s)
- Gregory R Steinberg
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada.
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada.
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada.
| | - D Grahame Hardie
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Dundee, UK.
| |
Collapse
|
29
|
Wang D, Zhang M, Xu J, Yang J. Uncarboxylated Osteocalcin Decreases SCD1 by Activating AMPK to Alleviate Hepatocyte Lipid Accumulation. Molecules 2023; 28:molecules28073121. [PMID: 37049884 PMCID: PMC10095730 DOI: 10.3390/molecules28073121] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/19/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
Uncarboxylated osteocalcin (GluOC), a small-molecule protein specifically synthesized and secreted by osteoblasts, is important in the regulation of energy metabolism. In our previous study, GluOC was shown to be effective in ameliorating dyslipidemia and hepatic steatosis in KKAy mice. However, the underlying mechanism of GluOC action on hepatocytes has not been well validated. In this study, oleic acid/palmitic acid (OA/PA)-induced HepG2 and NCTC 1469 cells were used as non-alcoholic fatty liver disease (NAFLD) cell models, and triacylglycerol (TG) levels were measured by oil red O staining, Nile Red staining, and ELISA. The fatty acid synthesis-related protein expression was detected by real-time quantitative polymerase chain reaction, Western blotting, and immunofluorescence. The results show that GluOC reduced triglyceride levels, and decreased the expression of sterol regulatory element-binding protein-1c (SREBP-1c) and stearyl-coenzyme A desaturase 1 (SCD1). si-SCD1 mimicked the lipid accumulation-reducing effect of GluOC, while overexpression of SCD1 attenuated the effect of GluOC. In addition, GluOC activated AMP-activated protein kinase (AMPK) phosphorylation to affect lipid metabolism in hepatocytes. Overall, the results of this study suggest that GluOC decreases SCD1 by activating AMPK to alleviate hepatocyte lipid accumulation, which provides a new target for improving NAFLD in further research.
Collapse
|
30
|
Tian R, Yang J, Wang X, Liu S, Dong R, Wang Z, Yang Z, Zhang Y, Cai Z, Yang H, Hu Y, She ZG, Li H, Zhou J, Zhang XJ. Honokiol acts as an AMPK complex agonist therapeutic in non-alcoholic fatty liver disease and metabolic syndrome. Chin Med 2023; 18:30. [PMID: 36932412 PMCID: PMC10024454 DOI: 10.1186/s13020-023-00729-5] [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: 12/11/2022] [Accepted: 02/15/2023] [Indexed: 03/19/2023] Open
Abstract
BACKGROUND Non-alcoholic fatty liver (NAFLD) and its related metabolic syndrome have become major threats to human health, but there is still a need for effective and safe drugs to treat these conditions. Here we aimed to identify potential drug candidates for NAFLD and the underlying molecular mechanisms. METHODS A drug repositioning strategy was used to screen an FDA-approved drug library with approximately 3000 compounds in an in vitro hepatocyte model of lipid accumulation, with honokiol identified as an effective anti-NAFLD candidate. We systematically examined the therapeutic effect of honokiol in NAFLD and metabolic syndrome in multiple in vitro and in vivo models. Transcriptomic examination and biotin-streptavidin binding assays were used to explore the underlying molecular mechanisms, confirmed by rescue experiments. RESULTS Honokiol significantly inhibited metabolic syndrome and NAFLD progression as evidenced by improved hepatic steatosis, liver fibrosis, adipose inflammation, and insulin resistance. Mechanistically, the beneficial effects of honokiol were largely through AMPK activation. Rather than acting on the classical upstream regulators of AMPK, honokiol directly bound to the AMPKγ1 subunit to robustly activate AMPK signaling. Mutation of honokiol-binding sites of AMPKγ1 largely abolished the protective capacity of honokiol against NAFLD. CONCLUSION These findings clearly demonstrate the beneficial effects of honokiol in multiple models and reveal a previously unappreciated signaling mechanism of honokiol in NAFLD and metabolic syndrome. This study also provides new insights into metabolic disease treatment by targeting AMPKγ1 subunit-mediated signaling activation.
Collapse
Affiliation(s)
- Ruifeng Tian
- Department of Cardiology,Renmin Hospital; School of Basic Medical Science, Wuhan University, Wuhan, 430060, China.,Institute of Model Animal of Wuhan University, Wuhan, 430071, China
| | - Jinjie Yang
- Department of Cardiology,Renmin Hospital; School of Basic Medical Science, Wuhan University, Wuhan, 430060, China.,Institute of Model Animal of Wuhan University, Wuhan, 430071, China
| | - Xiaoming Wang
- Department of Cardiology,Renmin Hospital; School of Basic Medical Science, Wuhan University, Wuhan, 430060, China.,Institute of Model Animal of Wuhan University, Wuhan, 430071, China
| | - Shuaiyang Liu
- Department of Cardiology,Renmin Hospital; School of Basic Medical Science, Wuhan University, Wuhan, 430060, China.,Institute of Model Animal of Wuhan University, Wuhan, 430071, China
| | - Ruixiang Dong
- Department of Cardiology,Renmin Hospital; School of Basic Medical Science, Wuhan University, Wuhan, 430060, China.,Institute of Model Animal of Wuhan University, Wuhan, 430071, China
| | - Zhenya Wang
- Department of Cardiology,Renmin Hospital; School of Basic Medical Science, Wuhan University, Wuhan, 430060, China.,Institute of Model Animal of Wuhan University, Wuhan, 430071, China
| | - Zifeng Yang
- Department of Cardiology,Renmin Hospital; School of Basic Medical Science, Wuhan University, Wuhan, 430060, China.,Institute of Model Animal of Wuhan University, Wuhan, 430071, China
| | - Yingping Zhang
- School of Pharmacy, Bengbu Medical College, Bengbu, 233030, China
| | - Zhiwei Cai
- Institute of Model Animal of Wuhan University, Wuhan, 430071, China
| | - Hailong Yang
- Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, 341000, China.,Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, 341000, China
| | - Yufeng Hu
- Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, 341000, China.,Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, 341000, China
| | - Zhi-Gang She
- Department of Cardiology,Renmin Hospital; School of Basic Medical Science, Wuhan University, Wuhan, 430060, China.,Institute of Model Animal of Wuhan University, Wuhan, 430071, China
| | - Hongliang Li
- Department of Cardiology,Renmin Hospital; School of Basic Medical Science, Wuhan University, Wuhan, 430060, China. .,Institute of Model Animal of Wuhan University, Wuhan, 430071, China. .,Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, 341000, China. .,Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, 341000, China. .,Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Junjie Zhou
- Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, 341000, China. .,Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, 341000, China.
| | - Xiao-Jing Zhang
- Department of Cardiology,Renmin Hospital; School of Basic Medical Science, Wuhan University, Wuhan, 430060, China. .,Institute of Model Animal of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
31
|
Yang Y, Reid MA, Hanse EA, Li H, Li Y, Ruiz BI, Fan Q, Kong M. SAPS3 subunit of protein phosphatase 6 is an AMPK inhibitor and controls metabolic homeostasis upon dietary challenge in male mice. Nat Commun 2023; 14:1368. [PMID: 36914647 PMCID: PMC10011557 DOI: 10.1038/s41467-023-36809-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 02/17/2023] [Indexed: 03/16/2023] Open
Abstract
Inhibition of AMPK is tightly associated with metabolic perturbations upon over nutrition, yet the molecular mechanisms underlying are not clear. Here, we demonstrate the serine/threonine-protein phosphatase 6 regulatory subunit 3, SAPS3, is a negative regulator of AMPK. SAPS3 is induced under high fat diet (HFD) and recruits the PP6 catalytic subunit to deactivate phosphorylated-AMPK, thereby inhibiting AMPK-controlled metabolic pathways. Either whole-body or liver-specific deletion of SAPS3 protects male mice against HFD-induced detrimental consequences and reverses HFD-induced metabolic and transcriptional alterations while loss of SAPS3 has no effects on mice under balanced diets. Furthermore, genetic inhibition of AMPK is sufficient to block the protective phenotype in SAPS3 knockout mice under HFD. Together, our results reveal that SAPS3 is a negative regulator of AMPK and suppression of SAPS3 functions as a guardian when metabolism is perturbed and represents a potential therapeutic strategy to treat metabolic syndromes.
Collapse
Affiliation(s)
- Ying Yang
- Department of Molecular Biology and Biochemistry; School of Biological Sciences, University of California, Irvine, Irvine, CA, 92697, USA
| | - Michael A Reid
- Department of Cancer Biology, Beckman Research Institute of City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Eric A Hanse
- Department of Molecular Biology and Biochemistry; School of Biological Sciences, University of California, Irvine, Irvine, CA, 92697, USA
| | - Haiqing Li
- Integrative Genomics Core, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Yuanding Li
- Department of Molecular Biology and Biochemistry; School of Biological Sciences, University of California, Irvine, Irvine, CA, 92697, USA
| | - Bryan I Ruiz
- Department of Molecular Biology and Biochemistry; School of Biological Sciences, University of California, Irvine, Irvine, CA, 92697, USA
| | - Qi Fan
- Department of Molecular Biology and Biochemistry; School of Biological Sciences, University of California, Irvine, Irvine, CA, 92697, USA
| | - Mei Kong
- Department of Molecular Biology and Biochemistry; School of Biological Sciences, University of California, Irvine, Irvine, CA, 92697, USA.
| |
Collapse
|
32
|
Wang G, Ge L, Liu T, Zheng Z, Chen L. The therapeutic potential of arctigenin against multiple human diseases: A mechanistic review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 110:154647. [PMID: 36628833 DOI: 10.1016/j.phymed.2023.154647] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 12/21/2022] [Accepted: 01/01/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND Arctigenin (ATG), a dibenzyl butyrolactone lignan compound, is one of the major bioactive components from the medicinal plant Arctium lappa. ATG possesses remarkable therapeutic potential against a wide range of human diseases, such as cancers, immune disorders and chronical diseases. The molecular mechanisms behind the biological effects of ATG have been intensively studied. PURPOSE This review aims to systematically summarize the updated knowledge of the proteins and signaling pathways behind the curative property of ATG, and further analyze the potential connections between them. METHOD SciFinder, Pubmed, Web of Science and Cochrane Library databases were queried for publications reporting the therapeutic properties of ATG. "Arctigenin", "disease", "cancer", "inflammation", "organ damage", "infection", "toxicity" and "pharmacokinetics" were used as the searching titles. RESULT 625 publications were identified and 95 met the inclusion criteria and exclusion criteria. 42 studies described the molecular mechanisms implicated in ATG treatments. Several proteins including phosphodiesterase subtype 4D (PDE4D), estrogen receptor (ER) β, protein phosphatase 2A (PP2A), phosphoinositide 3-kinase (PI3K) and transmembrane protein 16A (TMEM16A) are targeted by ATG in different settings. The frequently described signaling pathways are TLR4/NF-κB, PI3K/AKT/mTOR, AMP-activated protein kinase (AMPK) and nuclear factor erythroid 2-related factor 2 (Nrf-2) signalings. CONCLUSION Inhibition of PI3K/AKT pathway and activation of AMPK signaling play the pivotal roles in the therapeutic effects of ATG. PI3K/AKT and AMPK signaling widely link to other signaling pathways, modulating various biological processes such as anti-inflammation, anti-oxidative stress, anti-fibrosis, anti-ER stress, anti-steatosis and pro-apoptosis, which constitute the curative mechanisms of ATG against multiple human diseases.
Collapse
Affiliation(s)
- Guanming Wang
- School of Nursing, Fujian University of Traditional Chinese Medicine, Fuzhou, 350108, China.
| | - Li Ge
- School of Nursing, Fujian University of Traditional Chinese Medicine, Fuzhou, 350108, China
| | - Tongyu Liu
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Zhihui Zheng
- School of Nursing, Fujian University of Traditional Chinese Medicine, Fuzhou, 350108, China
| | - Lijun Chen
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China.
| |
Collapse
|
33
|
Tian H, Fang Y, Liu W, Wang J, Zhao J, Tang H, Yin Y, Hu Y, Peng J. Inhibition on XBP1s-driven lipogenesis by Qushi Huayu Decoction contributes to amelioration of hepatic steatosis induced by fructose. JOURNAL OF ETHNOPHARMACOLOGY 2023; 301:115806. [PMID: 36216198 DOI: 10.1016/j.jep.2022.115806] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/29/2022] [Accepted: 10/03/2022] [Indexed: 06/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Qushi Huayu Decoction (QHD) is a traditional Chinese medicine formula consisting of five herbs, which has been used for non-alcoholic fatty liver disease (NAFLD) treatment in clinic for decades in China and validated in several NAFLD animal models. The hepatic de novo lipogenesis (DNL) is enhanced greatly to contribute to steatosis in NAFLD. The spliced form of X-box binding protein 1 (XBP1s) initiates DNL independently of sterol regulatory element-binding protein (SREBP) and carbohydrate-responsive element-binding protein (ChREBP). AIM OF THE STUDY To disclose the mechanism of inhibition on hepatic DNL by QHD and the responsible compounds. METHODS The effects of QHD on hepatic DNL were evaluated in mice induced by high-fructose diet (HFru). The effects of the serum-absorbed compounds of QHD on XBP1s were evaluated in HepG2 cells induced by tunicamycin. Hepatic histology, triglyceride (TG) and nonesterified fatty acids were observed. Hepatic apolipoprotein B100 and very low-density lipoprotein were measured to reflect lipid out-transport. The mRNA expression of XBP1s and its target genes were detected by real-time polymerase chain reaction. The protein expression of TG synthetases and DNL enzymes, and inositol requirement enzyme 1 alpha (IRE1α), phosphorylated IRE1α and XBP1s were detected in liver tissue and HepG2 cells by western-blot. The binding activity of SREBP1, protein expression of ChREBP and XBP1s were detected in the nuclear extracts of liver tissue. RESULTS Dynamical observing suggested feeding with HFru for 2 weeks was sufficient to induce hepatic lipogenesis and XBP1s. QHD ameliorated liver steatosis without enhancing out-transport of lipids, accompanied with more inhibitory effects on DNL enzymes than TG synthetases. QHD inhibits the nuclear XBP1s without affecting ChREBP and SREBP1. In QHD, chlorogenic acid, geniposide and polydatin inhibit lipogenesis initiated by XPB1s. CONCLUSION QHD probably decreases hepatic DNL by inhibiting XBP1s independent of SREBP1 and ChREBP. Chlorogenic acid, geniposide and polydatin are the potential responsible compounds.
Collapse
Affiliation(s)
- Huajie Tian
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528, Zhangheng Road, Shanghai, China
| | - Yi Fang
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528, Zhangheng Road, Shanghai, China
| | - Wei Liu
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528, Zhangheng Road, Shanghai, China
| | - Jun Wang
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528, Zhangheng Road, Shanghai, China
| | - Jianan Zhao
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528, Zhangheng Road, Shanghai, China
| | - Hao Tang
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528, Zhangheng Road, Shanghai, China
| | - Yixiao Yin
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528, Zhangheng Road, Shanghai, China
| | - Yiyang Hu
- Institute of Clinical Pharmacology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528, Zhangheng Road, Shanghai, China; Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, 528, Zhangheng Road, Shanghai, China.
| | - Jinghua Peng
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528, Zhangheng Road, Shanghai, China; Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, 528, Zhangheng Road, Shanghai, China; Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, 528, Zhangheng Road, Shanghai, China.
| |
Collapse
|
34
|
AMPK inhibits liver gluconeogenesis: fact or fiction? Biochem J 2023; 480:105-125. [PMID: 36637190 DOI: 10.1042/bcj20220582] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/21/2022] [Accepted: 01/04/2023] [Indexed: 01/14/2023]
Abstract
Is there a role for AMPK in the control of hepatic gluconeogenesis and could targeting AMPK in liver be a viable strategy for treating type 2 diabetes? These are frequently asked questions this review tries to answer. After describing properties of AMPK and different small-molecule AMPK activators, we briefly review the various mechanisms for controlling hepatic glucose production, mainly via gluconeogenesis. The different experimental and genetic models that have been used to draw conclusions about the role of AMPK in the control of liver gluconeogenesis are critically discussed. The effects of several anti-diabetic drugs, particularly metformin, on hepatic gluconeogenesis are also considered. We conclude that the main effect of AMPK activation pertinent to the control of hepatic gluconeogenesis is to antagonize glucagon signalling in the short-term and, in the long-term, to improve insulin sensitivity by reducing hepatic lipid content.
Collapse
|
35
|
Blocking AMPK β1 myristoylation enhances AMPK activity and protects mice from high-fat diet-induced obesity and hepatic steatosis. Cell Rep 2022; 41:111862. [PMID: 36543129 DOI: 10.1016/j.celrep.2022.111862] [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: 03/11/2022] [Revised: 10/07/2022] [Accepted: 11/30/2022] [Indexed: 12/24/2022] Open
Abstract
AMP-activated protein kinase (AMPK) is a master regulator of cellular energy homeostasis and a therapeutic target for metabolic diseases. Co/post-translational N-myristoylation of glycine-2 (Gly2) of the AMPK β subunit has been suggested to regulate the distribution of the kinase between the cytosol and membranes through a "myristoyl switch" mechanism. However, the relevance of AMPK myristoylation for metabolic signaling in cells and in vivo is unclear. Here, we generated knockin mice with a Gly2-to-alanine point mutation of AMPKβ1 (β1-G2A). We demonstrate that non-myristoylated AMPKβ1 has reduced stability but is associated with increased kinase activity and phosphorylation of the Thr172 activation site in the AMPK α subunit. Using proximity ligation assays, we show that loss of β1 myristoylation impedes colocalization of the phosphatase PPM1A/B with AMPK in cells. Mice carrying the β1-G2A mutation have improved metabolic health with reduced adiposity, hepatic lipid accumulation, and insulin resistance under conditions of high-fat diet-induced obesity.
Collapse
|
36
|
Alshuniaber MA, Alshammari GM, Eleawa SM, Yagoub AEA, Al-Khalifah AS, Alhussain MH, Al-Harbi LN, Yahya MA. Camel milk protein hydrosylate alleviates hepatic steatosis and hypertension in high fructose-fed rats. PHARMACEUTICAL BIOLOGY 2022; 60:1137-1147. [PMID: 35672152 PMCID: PMC9176680 DOI: 10.1080/13880209.2022.2079678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 04/19/2022] [Accepted: 05/14/2022] [Indexed: 06/15/2023]
Abstract
CONTEXT Camel milk is used in traditional medicine to treat diabetes mellitus hypertension and other metabolic disorders. OBJECTIVE This study evaluated the antisteatotic and antihypertensive effects of camel milk protein hydrolysate (CMH) in high fructose (HF)-fed rats and compared it with the effects afforded by the intact camel milk protein extract (ICM). MATERIALS AND METHODS Adult male Wistar rats were divided into 6 groups (n = 8 each) as 1) control, 2) ICM (1000 mg/kg), 3) CMH (1000 mg/kg), 4) HF (15% in drinking water), 5) HF (15%) + ICM (1000 mg/kg), and 6) HF (15%) + CMH (1000 mg/kg). All treatments were given orally for 21 weeks, daily. RESULTS Both ICM and CMH reduced fasting glucose and insulin levels, serum and hepatic levels of cholesterol and triglycerides, and serum levels of ALT and AST, angiotensin II, ACE, endothelin-1, and uric acid in HF-fed rats. In addition, both ICM and CMH reduced hepatic fat deposition in the hepatocytes and reduced hepatocyte damage. This was associated with an increase in the hepatic activity of AMPK, higher PPARα mRNA, reduced expression of fructokinase C, SREBP1, SREBP2, fatty acid synthase, and HMG-CoA-reductase. Both treatments lowered systolic and diastolic blood pressure. However, the effects of CMH on all these parameters were greater as compared to ICM. DISCUSSION AND CONCLUSIONS The findings of this study encourage the use of CMH in a large-scale population and clinical studies to treat metabolic steatosis and hypertension.
Collapse
Affiliation(s)
- Mohammad A. Alshuniaber
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Ghedeir M. Alshammari
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Samy M. Eleawa
- College of Health Sciences, Applied Medical Sciences Department, PAAET, Safat, Kuwait
| | - Abu ElGasim A. Yagoub
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Abdullrahman S. Al-Khalifah
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Maha H. Alhussain
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Laila Naif Al-Harbi
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Mohammed Abdo Yahya
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
37
|
Jung TW, Kim H, Park SY, Cho W, Oh H, Lee HJ, Abd El-Aty AM, Hacimuftuoglu A, Jeong JH. Stachydrine alleviates lipid-induced skeletal muscle insulin resistance via AMPK/HO-1-mediated suppression of inflammation and endoplasmic reticulum stress. J Endocrinol Invest 2022; 45:2181-2191. [PMID: 35834165 DOI: 10.1007/s40618-022-01866-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 07/06/2022] [Indexed: 10/17/2022]
Abstract
OBJECTIVE Insulin resistance develops due to skeletal muscle inflammation and endoplasmic reticulum (ER) stress. Stachydrine (STA), extracted from Leonurus heterophyllus, has been shown to suppress proliferation and induce apoptosis in breast cancer cells and exert anti-inflammatory properties in the brain, heart, and liver. However, the roles of STA in insulin signaling in skeletal muscle remain unclear. Herein, we examined the impacts of STA on insulin signaling in skeletal muscle under hyperlipidemic conditions and its related molecular mechanisms. METHODS Various protein expression levels were determined by Western blotting. Levels of mouse serum cytokines were measured by ELISA. RESULTS We found that STA-ameliorated inflammation and ER stress, leading to attenuation of insulin resistance in palmitate-treated C2C12 myocytes. STA dose-dependently enhanced AMPK phosphorylation and HO-1 expression. Administration of STA attenuated not only insulin resistance but also inflammation and ER stress in the skeletal muscle of high-fat diet (HFD)-fed mice. Additionally, STA-ameliorated glucose tolerance and insulin sensitivity, as well as serum TNFα and MCP-1, in mice fed a HFD. Small interfering (si) RNA-associated suppression of AMPK or HO-1 expression abolished the effects of STA in C2C12 myocytes. CONCLUSIONS These results suggest that STA activates AMPK/HO-1 signaling, resulting in reduced inflammation and ER stress, thereby improving skeletal muscle insulin resistance. Using STA as a natural ingredient, this research successfully treated insulin resistance and type 2 diabetes.
Collapse
Affiliation(s)
- T W Jung
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - H Kim
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - S Y Park
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, Republic of Korea
| | - W Cho
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - H Oh
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - H J Lee
- Department of Anatomy and Cell Biology, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Korea
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, Republic of Korea
| | - A M Abd El-Aty
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
- Department of Medical Pharmacology, Medical Faculty, Ataturk University, 25240, Erzurum, Türkiye
| | - A Hacimuftuoglu
- Department of Medical Pharmacology, Medical Faculty, Ataturk University, 25240, Erzurum, Türkiye
| | - J H Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea.
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, Republic of Korea.
| |
Collapse
|
38
|
Zhang CS, Li M, Wang Y, Li X, Zong Y, Long S, Zhang M, Feng JW, Wei X, Liu YH, Zhang B, Wu J, Zhang C, Lian W, Ma T, Tian X, Qu Q, Yu Y, Xiong J, Liu DT, Wu Z, Zhu M, Xie C, Wu Y, Xu Z, Yang C, Chen J, Huang G, He Q, Huang X, Zhang L, Sun X, Liu Q, Ghafoor A, Gui F, Zheng K, Wang W, Wang ZC, Yu Y, Zhao Q, Lin SY, Wang ZX, Piao HL, Deng X, Lin SC. The aldolase inhibitor aldometanib mimics glucose starvation to activate lysosomal AMPK. Nat Metab 2022; 4:1369-1401. [PMID: 36217034 PMCID: PMC9584815 DOI: 10.1038/s42255-022-00640-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 08/16/2022] [Indexed: 01/20/2023]
Abstract
The activity of 5'-adenosine monophosphate-activated protein kinase (AMPK) is inversely correlated with the cellular availability of glucose. When glucose levels are low, the glycolytic enzyme aldolase is not bound to fructose-1,6-bisphosphate (FBP) and, instead, signals to activate lysosomal AMPK. Here, we show that blocking FBP binding to aldolase with the small molecule aldometanib selectively activates the lysosomal pool of AMPK and has beneficial metabolic effects in rodents. We identify aldometanib in a screen for aldolase inhibitors and show that it prevents FBP from binding to v-ATPase-associated aldolase and activates lysosomal AMPK, thereby mimicking a cellular state of glucose starvation. In male mice, aldometanib elicits an insulin-independent glucose-lowering effect, without causing hypoglycaemia. Aldometanib also alleviates fatty liver and nonalcoholic steatohepatitis in obese male rodents. Moreover, aldometanib extends lifespan and healthspan in both Caenorhabditis elegans and mice. Taken together, aldometanib mimics and adopts the lysosomal AMPK activation pathway associated with glucose starvation to exert physiological roles, and might have potential as a therapeutic for metabolic disorders in humans.
Collapse
Affiliation(s)
- Chen-Song Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, China
| | - Mengqi Li
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, China
| | - Yu Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, China
| | - Xiaoyang Li
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, China
| | - Yue Zong
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, China
| | - Shating Long
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, China
| | - Mingliang Zhang
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jin-Wei Feng
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, China
| | - Xiaoyan Wei
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, China
| | - Yan-Hui Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, China
| | - Baoding Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, China
| | - Jianfeng Wu
- Laboratory Animal Research Centre, Xiamen University, Fujian, China
| | - Cixiong Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, China
| | - Wenhua Lian
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, China
| | - Teng Ma
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, China
| | - Xiao Tian
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, China
| | - Qi Qu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, China
| | - Yaxin Yu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, China
| | - Jinye Xiong
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, China
| | - Dong-Tai Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, China
| | - Zhenhua Wu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, China
| | - Mingxia Zhu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, China
| | - Changchuan Xie
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, China
| | - Yaying Wu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, China
| | - Zheni Xu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, China
| | - Chunyan Yang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, China
| | - Junjie Chen
- Analysis and Measurement Centre, School of Pharmaceutical Sciences, Xiamen University, Fujian, China
| | - Guohong Huang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, China
| | - Qingxia He
- Key Laboratory of Ministry of Education for Protein Science, School of Life Sciences, Tsinghua University, Beijing, China
| | - Xi Huang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, China
| | - Lei Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, China
| | - Xiufeng Sun
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, China
| | - Qingfeng Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, China
| | - Abdul Ghafoor
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, China
| | - Fu Gui
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, China
| | - Kaili Zheng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Fujian, China
| | - Wen Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Liaoning, China
| | - Zhi-Chao Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Liaoning, China
| | - Yong Yu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, China
| | - Qingliang Zhao
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Fujian, China
| | - Shu-Yong Lin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, China
| | - Zhi-Xin Wang
- Key Laboratory of Ministry of Education for Protein Science, School of Life Sciences, Tsinghua University, Beijing, China
| | - Hai-Long Piao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Liaoning, China
| | - Xianming Deng
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, China.
| | - Sheng-Cai Lin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, China.
| |
Collapse
|
39
|
Song L, Wang L, Hou Y, Zhou J, Chen C, Ye X, Dong W, Gao H, Liu Y, Qiao G, Pan T, Chen Q, Cao Y, Hu F, Rao Z, Chen Y, Han Y, Zheng M, Luo Y, Li X, Chen Y, Huang Z. FGF4 protects the liver from nonalcoholic fatty liver disease by activating the AMP-activated protein kinase-Caspase 6 signal axis. Hepatology 2022; 76:1105-1120. [PMID: 35152446 DOI: 10.1002/hep.32404] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 02/04/2022] [Accepted: 02/05/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS NAFLD represents an increasing health problem in association with obesity and diabetes with no effective pharmacotherapies. Growing evidence suggests that several FGFs play important roles in diverse aspects of liver pathophysiology. Here, we report a previously unappreciated role of FGF4 in the liver. APPROACH AND RESULTS Expression of hepatic FGF4 is inversely associated with NAFLD pathological grades in both human patients and mouse models. Loss of hepatic Fgf4 aggravates hepatic steatosis and liver damage resulted from an obesogenic high-fat diet. By contrast, pharmacological administration of recombinant FGF4 mitigates hepatic steatosis, inflammation, liver damage, and fibrogenic markers in mouse livers induced to develop NAFLD and NASH under dietary challenges. Such beneficial effects of FGF4 are mediated predominantly by activating hepatic FGF receptor (FGFR) 4, which activates a downstream Ca2+ -Ca2+ /calmodulin-dependent protein kinase kinase beta-dependent AMP-activated protein kinase (AMPK)-Caspase 6 signal axis, leading to enhanced fatty acid oxidation, reduced hepatocellular apoptosis, and mitigation of liver damage. CONCLUSIONS Our study identifies FGF4 as a stress-responsive regulator of liver pathophysiology that acts through an FGFR4-AMPK-Caspase 6 signal pathway, shedding light on strategies for treating NAFLD and associated liver pathologies.
Collapse
Affiliation(s)
- Lintao Song
- Department of Infectious DiseasesZhejiang Provincial Key Laboratory of Liver DiseasesThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
- School of Pharmaceutical SciencesInstitute of AgingWenzhou Medical UniversityWenzhouZhejiangChina
| | - Luyao Wang
- School of Pharmaceutical SciencesInstitute of AgingWenzhou Medical UniversityWenzhouZhejiangChina
| | - Yushu Hou
- School of Pharmaceutical SciencesInstitute of AgingWenzhou Medical UniversityWenzhouZhejiangChina
| | - Jie Zhou
- School of Pharmaceutical SciencesInstitute of AgingWenzhou Medical UniversityWenzhouZhejiangChina
| | - Chuchu Chen
- School of Pharmaceutical SciencesInstitute of AgingWenzhou Medical UniversityWenzhouZhejiangChina
| | - Xianxi Ye
- School of Pharmaceutical SciencesInstitute of AgingWenzhou Medical UniversityWenzhouZhejiangChina
| | - Wenliya Dong
- School of Pharmaceutical SciencesInstitute of AgingWenzhou Medical UniversityWenzhouZhejiangChina
| | - Huan Gao
- School of Pharmaceutical SciencesInstitute of AgingWenzhou Medical UniversityWenzhouZhejiangChina
| | - Yi Liu
- School of Pharmaceutical SciencesInstitute of AgingWenzhou Medical UniversityWenzhouZhejiangChina
| | - Guanting Qiao
- School of Pharmaceutical SciencesInstitute of AgingWenzhou Medical UniversityWenzhouZhejiangChina
| | - Tongtong Pan
- Department of Infectious DiseasesZhejiang Provincial Key Laboratory of Liver DiseasesThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| | - Qiong Chen
- School of Pharmaceutical SciencesInstitute of AgingWenzhou Medical UniversityWenzhouZhejiangChina
| | - Yu Cao
- School of Pharmaceutical SciencesInstitute of AgingWenzhou Medical UniversityWenzhouZhejiangChina
| | - Fengjiao Hu
- Medical Science Research CenterZhongnan HospitalWuhan UniversityWuhanChina
| | - Zhiheng Rao
- School of Pharmaceutical SciencesInstitute of AgingWenzhou Medical UniversityWenzhouZhejiangChina
| | - Yajing Chen
- School of Pharmaceutical SciencesInstitute of AgingWenzhou Medical UniversityWenzhouZhejiangChina
| | - Yu Han
- Department of Infectious DiseasesZhejiang Provincial Key Laboratory of Liver DiseasesThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| | - Minghua Zheng
- NAFLD Research CenterDepartment of HepatologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| | - Yongde Luo
- School of Pharmaceutical SciencesInstitute of AgingWenzhou Medical UniversityWenzhouZhejiangChina
- NAFLD Research CenterDepartment of HepatologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| | - Xiaokun Li
- School of Pharmaceutical SciencesInstitute of AgingWenzhou Medical UniversityWenzhouZhejiangChina
| | - Yongping Chen
- Department of Infectious DiseasesZhejiang Provincial Key Laboratory of Liver DiseasesThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| | - Zhifeng Huang
- Department of Infectious DiseasesZhejiang Provincial Key Laboratory of Liver DiseasesThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
- School of Pharmaceutical SciencesInstitute of AgingWenzhou Medical UniversityWenzhouZhejiangChina
| |
Collapse
|
40
|
Wang L, Dong W, Gao H, Chen C, Liang S, Ye X, Liu Y, Hou Y, Fan L, Pan T, Wang Z, Chen Y, Luo Y, Song L. A non-mitogenic FGF4 analog alleviates non-alcoholic steatohepatitis through an AMPK-dependent pathway. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166560. [PMID: 36167161 DOI: 10.1016/j.bbadis.2022.166560] [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: 07/23/2022] [Revised: 09/16/2022] [Accepted: 09/22/2022] [Indexed: 10/14/2022]
Abstract
BACKGROUND & AIMS Non-alcoholic fatty liver disease (NAFLD) has emerged as a major liver disease increasingly in association with non-alcoholic steatohepatitis (NASH), cirrhosis and hepatocellular carcinoma (HCC). However, there are currently no approved therapies for treating NAFLD and NASH. Fibroblast growth factor 4 (FGF4) has recently been shown as a promising drug candidate for several metabolic diseases. METHODS Mice fed a high-fat diet with high fructose/glucose drinking water (HF/HFG, Western-like diet) for 21 weeks were intraperitoneally injected with non-mitogenic recombinant FGF4△NT (rFGF4△NT, 1.0 mg/kg body weight) every other day for 8 weeks. Primary mouse hepatocytes cultured in medium containing high glucose/palmitic acid (HG/PA) or TNFα/cyclohexane (TNFα/CHX) were treated with 1.0 μg/ml rFGF4△NT. Changes in parameters for histopathology, lipid metabolism, inflammation, hepatocellular apoptosis and fibrosis were determined. The Caspase6 activity and AMPK pathway were assessed. RESULTS Administration of rFGF4△NT significantly attenuated the Western-like diet-induced hepatic steatosis, inflammation, liver injury and fibrosis in mice. rFGF4△NT treatment reduced fatty acid-induced lipid accumulation and lipotoxicity-induced hepatocyte apoptosis, which were associated with inhibition of Caspase6 cleavage and activation. Inhibition of AMP-activated protein kinase (AMPK) by Compound C or deficiency of Ampk abrogated rFGF4△NT-induced hepatoprotection in primary hepatocytes and in mice with NASH. CONCLUSION rFGF4△NT exerts significant protective effects on NASH via an AMPK-dependent signaling pathway. Our study indicates that FGF4 analogs may have therapeutic potential for the Western-like diet induced NASH.
Collapse
Affiliation(s)
- Luyao Wang
- The First Affiliated Hospital and School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Wenliya Dong
- The First Affiliated Hospital and School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Huan Gao
- The First Affiliated Hospital and School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Chuchu Chen
- The First Affiliated Hospital and School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Siyu Liang
- The 2nd Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xianxi Ye
- The First Affiliated Hospital and School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yi Liu
- The First Affiliated Hospital and School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yushu Hou
- The First Affiliated Hospital and School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Lei Fan
- The First Affiliated Hospital and School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Clinical Pharmacy Research Center, Jinhua Hospital of Zhejiang University and Jinhua Municipal Central Hospital, Jinhua, Zhejiang 321000, China
| | - Tongtong Pan
- The First Affiliated Hospital and School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zengshou Wang
- The 2nd Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Yongping Chen
- The First Affiliated Hospital and School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Yongde Luo
- The First Affiliated Hospital and School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Lintao Song
- Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| |
Collapse
|
41
|
Sun H, Seok S, Jung H, Kemper B, Kemper JK. Obesity-induced miR-802 directly targets AMPK and promotes nonalcoholic steatohepatitis in mice. Mol Metab 2022; 66:101603. [PMID: 36126896 PMCID: PMC9515436 DOI: 10.1016/j.molmet.2022.101603] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 09/13/2022] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVE Obesity-associated nonalcoholic fatty liver disease (NAFLD) is a leading cause of liver failure and death. However, the pathogenesis of NAFLD and its severe form, nonalcoholic steatohepatitis (NASH), is poorly understood. The energy sensor, AMP-activated protein kinase (AMPK), has decreased activity in obesity and NAFLD, but the mechanisms are unclear. Here, we examined whether obesity-induced miR-802 has a role in promoting NASH by targeting AMPK. We also investigated whether miR-802 and AMPK have roles in modulating beneficial therapeutic effects mediated by obeticholic acid (OCA), a promising clinical agent for NASH. METHODS Immunoblotting, luciferase assays, and RNA-protein interaction studies were performed to test whether miR-802 directly targets AMPK. The roles of miR-802 and AMPK in NASH were examined in mice fed a NASH-promoting diet. RESULTS Hepatic miR-802 and AMPK levels were inversely correlated in both NAFLD patients and obese mice. MicroRNA in silico analysis, together with biochemical studies in hepatic cells, suggested that miR-802 inhibits hepatic expression of AMPK by binding to the 3' untranslated regions of both human AMPKα1 and mouse Ampkβ1. In diet-induced NASH mice, OCA treatment reduced hepatic miR-802 levels and improved AMPK activity, ameliorating steatosis, inflammation, and apoptosis, but these OCA-mediated beneficial effects on NASH pathologies, particularly reducing apoptosis, were reversed by overexpression of miR-802 or downregulation of AMPK. CONCLUSIONS These results indicate that miR-802 inhibits AMPK by directly targeting Ampkβ1, promoting NAFLD/NASH in mice. The miR-802-AMPK axis that modulates OCA-mediated beneficial effects on NASH may represent a new therapeutic target.
Collapse
Affiliation(s)
| | | | | | | | - Jongsook Kim Kemper
- Corresponding author. Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, 407 S. Goodwin Avenue, Urbana, IL, 61801, USA
| |
Collapse
|
42
|
McGlone ER, Ansell TB, Dunsterville C, Song W, Carling D, Tomas A, Bloom SR, Sansom MSP, Tan T, Jones B. Hepatocyte cholesterol content modulates glucagon receptor signalling. Mol Metab 2022; 63:101530. [PMID: 35718339 PMCID: PMC9254120 DOI: 10.1016/j.molmet.2022.101530] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 06/13/2022] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE To determine whether glucagon receptor (GCGR) actions are modulated by cellular cholesterol levels. METHODS We determined the effects of experimental cholesterol depletion and loading on glucagon-mediated cAMP production, ligand internalisation and glucose production in human hepatoma cells, mouse and human hepatocytes. GCGR interactions with lipid bilayers were explored using coarse-grained molecular dynamic simulations. Glucagon responsiveness was measured in mice fed a high cholesterol diet with or without simvastatin to modulate hepatocyte cholesterol content. RESULTS GCGR cAMP signalling was reduced by higher cholesterol levels across different cellular models. Ex vivo glucagon-induced glucose output from mouse hepatocytes was enhanced by simvastatin treatment. Mice fed a high cholesterol diet had increased hepatic cholesterol and a blunted hyperglycaemic response to glucagon, both of which were partially reversed by simvastatin. Simulations identified likely membrane-exposed cholesterol binding sites on the GCGR, including a site where cholesterol is a putative negative allosteric modulator. CONCLUSIONS Our results indicate that cellular cholesterol content influences glucagon sensitivity and indicate a potential molecular basis for this phenomenon. This could be relevant to the pathogenesis of non-alcoholic fatty liver disease, which is associated with both hepatic cholesterol accumulation and glucagon resistance.
Collapse
Affiliation(s)
- Emma Rose McGlone
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London W12 0NN, United Kingdom; Department of Surgery and Cancer, Imperial College London, London W12 0NN, United Kingdom.
| | - T Bertie Ansell
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom.
| | - Cecilia Dunsterville
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London W12 0NN, United Kingdom.
| | - Wanling Song
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom.
| | - David Carling
- Cellular Stress Research Group, MRC London Institute of Medical Sciences, Imperial College London, London W12 0NN, United Kingdom.
| | - Alejandra Tomas
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London W12 0NN, United Kingdom.
| | - Stephen R Bloom
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London W12 0NN, United Kingdom.
| | - Mark S P Sansom
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom.
| | - Tricia Tan
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London W12 0NN, United Kingdom.
| | - Ben Jones
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London W12 0NN, United Kingdom.
| |
Collapse
|
43
|
Widjaja AA, Viswanathan S, Wei Ting JG, Tan J, Shekeran SG, Carling D, Lim WW, Cook SA. IL11 stimulates ERK/P90RSK to inhibit LKB1/AMPK and activate mTOR initiating a mesenchymal program in stromal, epithelial, and cancer cells. iScience 2022; 25:104806. [PMID: 35992082 PMCID: PMC9386112 DOI: 10.1016/j.isci.2022.104806] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 06/04/2022] [Accepted: 07/15/2022] [Indexed: 11/30/2022] Open
Abstract
IL11 initiates fibroblast activation but also causes epithelial cell dysfunction. The mechanisms underlying these processes are not known. We report that IL11-stimulated ERK/P90RSK activity causes the phosphorylation of LKB1 at S325 and S428, leading to its inactivation. This inhibits AMPK and activates mTOR across cell types. In stromal cells, IL11-stimulated ERK activity inhibits LKB1/AMPK which is associated with mTOR activation, ⍺SMA expression, and myofibroblast transformation. In hepatocytes and epithelial cells, IL11/ERK activity inhibits LKB1/AMPK leading to mTOR activation, SNAI1 expression, and cell dysfunction. Across cells, IL11-induced phenotypes were inhibited by metformin stimulated AMPK activation. In mice, genetic or pharmacologic manipulation of IL11 activity revealed a critical role of IL11/ERK signaling for LKB1/AMPK inhibition and mTOR activation in fatty liver disease. These data identify the IL11/mTOR axis as a signaling commonality in stromal, epithelial, and cancer cells and reveal a shared IL11-driven mesenchymal program across cell types.
Collapse
Affiliation(s)
- Anissa A Widjaja
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore 169857, Singapore
| | - Sivakumar Viswanathan
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore 169857, Singapore
| | - Joyce Goh Wei Ting
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore 169857, Singapore
| | - Jessie Tan
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore 169609, Singapore
| | - Shamini G Shekeran
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore 169857, Singapore
| | - David Carling
- MRC-London Institute of Medical Sciences, Hammersmith Hospital Campus, London W12 0NN, UK
| | - Wei-Wen Lim
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore 169857, Singapore.,National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore 169609, Singapore
| | - Stuart A Cook
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore 169857, Singapore.,National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore 169609, Singapore.,MRC-London Institute of Medical Sciences, Hammersmith Hospital Campus, London W12 0NN, UK
| |
Collapse
|
44
|
Lower serum copper concentrations are associated with higher prevalence of nonalcoholic steatohepatitis: a matched case-control study. Eur J Gastroenterol Hepatol 2022; 34:838-843. [PMID: 35694803 DOI: 10.1097/meg.0000000000002392] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
BACKGROUND AND AIM Copper is an essential trace element involved in oxidative stress reactions and energy metabolism. While nonalcoholic fatty liver disease (NAFLD) is closely related to metabolic dysfunction, the role of copper in the development of simple steatosis (NAFL) and nonalcoholic steatohepatitis (NASH) is still unclear. We aimed to compare serum copper levels between patients with simple steatosis and those with NASH. METHODS AND RESULTS We studied 102 patients with biopsy-proven NASH (cases) and 102 NAFL controls, who were matched for age, sex, and residential city. Multivariable conditional logistic analysis was performed to explore associations between serum copper levels and the presence of NASH. Serum copper levels were significantly lower in patients with NASH than in those with matched NAFL controls (15.53 ± 2.41 μmol/l vs. 16.34 ± 3.23 μmol/l; P = 0.029). This intergroup difference in serum copper levels was more pronounced in men than in women. The per unit, per SD, and per doubling of serum copper levels were associated, respectively, with an approximately 20, 40, and 90% decrease in risk of having NASH, even after adjustment for potential confounding factors. CONCLUSION Lower serum copper concentrations are significantly associated with higher prevalence of NASH among biopsied-proven NAFLD patients, particularly in men.
Collapse
|
45
|
Zhao G, Yang L, Zhong W, Hu Y, Tan Y, Ren Z, Ban Q, Yang CS, Wang Y, Wang Z. Polydatin, A Glycoside of Resveratrol, Is Better Than Resveratrol in Alleviating Non-alcoholic Fatty Liver Disease in Mice Fed a High-Fructose Diet. Front Nutr 2022; 9:857879. [PMID: 35651514 PMCID: PMC9149290 DOI: 10.3389/fnut.2022.857879] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 03/04/2022] [Indexed: 12/14/2022] Open
Abstract
Resveratrol (RES) is considered to be an activator of AMP-activated protein kinase (AMPK) with many reported health benefits. Polydatin (POD) is a natural precursor and glycosylated form of RES. The glycoside structure of POD alters the bioactivity. Overnutrition-stimulated reactive oxygen species (ROS) promote the AMPK suppression and metabolic dysregulation. The present work compared the effects of POD and RES in ameliorating energy homeostasis imbalance in mice fed a high-fructose diet and elucidated the underlying mechanisms of action. Our results showed that POD elevated the fecal levels of valeric acid and caproic acid via modification of gut microbiota, while RES did not significantly influence the levels of fecal short-chain fatty acids (SCFAs). Both POD and RES markedly decreased the oxidative stress and activated the AMPK signaling pathways in the liver. POD and RES exerted a similar effect in alleviating glucose dysmetabolism, but POD was more effective in ameliorating lipid dysmetabolism than RES. Furthermore, valeric acid and caproic acid alone can activate the AMPK and ameliorate hypercholesterolemia, and enhance the effects of POD on improving lipid metabolism in mice. Overall, for the first time, we demonstrated that POD administration elevated the fecal levels of valeric acid and caproic acid by modifying gut microbiota, thus promoting AMPK activation may be the underlying mechanism that POD is superior to RES in alleviating the lipid dysmetabolism. Our results suggest that POD may be an alternative for RES as an AMPK activator.
Collapse
Affiliation(s)
- Guangshan Zhao
- Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China.,Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, China.,School of Food Science and Technology, Henan Agricultural University, Zhengzhou, China.,Guangdong Province Key Laboratory of Bioengineering Medicine, Guangzhou, China.,Guangdong Provincial Biotechnology Drug and Engineering Technology Research Center, Guangzhou, China.,Guangzhou Jinan Biomedicine Research and Development Center Co., Ltd., Guangzhou, China
| | - Lian Yang
- Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Wenshen Zhong
- Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yuze Hu
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Yu Tan
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Zhe Ren
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, China.,Guangdong Provincial Biotechnology Drug and Engineering Technology Research Center, Guangzhou, China.,Guangzhou Jinan Biomedicine Research and Development Center Co., Ltd., Guangzhou, China
| | - Qiuyan Ban
- College of Horticulture, Henan Agricultural University, Zhengzhou, China
| | - Chung S Yang
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| | - Yifei Wang
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, China.,Guangdong Provincial Biotechnology Drug and Engineering Technology Research Center, Guangzhou, China.,Guangzhou Jinan Biomedicine Research and Development Center Co., Ltd., Guangzhou, China
| | - Zhiping Wang
- Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
46
|
Nguyen-Tu MS, Harris J, Martinez-Sanchez A, Chabosseau P, Hu M, Georgiadou E, Pollard A, Otero P, Lopez-Noriega L, Leclerc I, Sakamoto K, Schmoll D, Smith DM, Carling D, Rutter GA. Opposing effects on regulated insulin secretion of acute vs chronic stimulation of AMP-activated protein kinase. Diabetologia 2022; 65:997-1011. [PMID: 35294578 PMCID: PMC9076735 DOI: 10.1007/s00125-022-05673-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 12/13/2021] [Indexed: 11/26/2022]
Abstract
AIMS/HYPOTHESIS Although targeted in extrapancreatic tissues by several drugs used to treat type 2 diabetes, the role of AMP-activated protein kinase (AMPK) in the control of insulin secretion is still debatable. Previous studies have used pharmacological activators of limited selectivity and specificity, and none has examined in primary pancreatic beta cells the actions of the latest generation of highly potent and specific activators that act via the allosteric drug and metabolite (ADaM) site. METHODS AMPK was activated acutely in islets isolated from C57BL6/J mice, and in an EndoC-βH3 cell line, using three structurally distinct ADaM site activators (991, PF-06409577 and RA089), with varying selectivity for β1- vs β2-containing complexes. Mouse lines expressing a gain-of-function mutation in the γ1 AMPK subunit (D316a) were generated to examine the effects of chronic AMPK stimulation in the whole body, or selectively in the beta cell. RESULTS Acute (1.5 h) treatment of wild-type mouse islets with 991, PF-06409577 or RA089 robustly stimulated insulin secretion at high glucose concentrations (p<0.01, p<0.05 and p<0.001, respectively), despite a lowering of glucose-induced intracellular free Ca2+ dynamics in response to 991 (AUC, p<0.05) and to RA089 at the highest dose (25 μmol/l) at 5.59 min (p<0.05). Although abolished in the absence of AMPK, the effects of 991 were observed in the absence of the upstream kinase, liver kinase B1, further implicating 'amplifying' pathways. In marked contrast, chronic activation of AMPK, either globally or selectively in the beta cell, achieved using a gain-of-function mutant, impaired insulin release in vivo (p<0.05 at 15 min following i.p. injection of 3 mmol/l glucose) and in vitro (p<0.01 following incubation of islets with 17 mmol/l glucose), and lowered glucose tolerance (p<0.001). CONCLUSIONS/INTERPRETATION AMPK activation exerts complex, time-dependent effects on insulin secretion. These observations should inform the design and future clinical use of AMPK modulators.
Collapse
Affiliation(s)
- Marie-Sophie Nguyen-Tu
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Joseph Harris
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Aida Martinez-Sanchez
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Pauline Chabosseau
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Ming Hu
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Eleni Georgiadou
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Alice Pollard
- MRC- London Institute of Medical Sciences, Imperial College London, London, UK
- Structure Biophysics and Fragments, Discovery Sciences, AstraZeneca R&D, Cambridge, UK
| | - Pablo Otero
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Livia Lopez-Noriega
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Isabelle Leclerc
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Kei Sakamoto
- Novo Nordisk Center for Basic Metabolic Research, Copenhagen, Denmark
| | - Dieter Schmoll
- Sanofi-Aventis Deutschland GmbH, Frankfurt am Main, Germany
| | - David M Smith
- Emerging Innovations Unit, Discovery Sciences, AstraZeneca R&D , Cambridge, UK
| | - David Carling
- MRC- London Institute of Medical Sciences, Imperial College London, London, UK
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Republic of Singapore.
- CR-CHUM, University of Montréal, Montréal, QC, Canada.
| |
Collapse
|
47
|
Miyata N, Ito T, Nakashima M, Fujii S, Kuge O. Mitochondrial phosphatidylethanolamine synthesis affects mitochondrial energy metabolism and quiescence entry through attenuation of Snf1/AMPK signaling in yeast. FASEB J 2022; 36:e22355. [PMID: 35639425 DOI: 10.1096/fj.202101600rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 05/04/2022] [Accepted: 05/06/2022] [Indexed: 11/11/2022]
Abstract
The Ups2-Mdm35 complex mediates intramitochondrial phosphatidylserine (PS) transport to facilitate mitochondrial phosphatidylethanolamine (PE) synthesis. In the present study, we found that ups2∆ yeast showed increased mitochondrial ATP production and enhanced quiescence (G0) entry in the post-diauxic shift phase. Transcriptomic and biochemical analyses revealed that the depletion of Ups2 leads to overactivation of the yeast AMPK homolog Snf1. Inactivation of Snf1 by depletion of an Snf1-activating kinase, Sak1 canceled the changes in mitochondrial ATP production and quiescence entry observed in ups2∆ cells. Furthermore, among the factors regulated by Snf1, upregulation of pyruvate carboxylase, Pyc1 and downregulation of acetyl-CoA carboxylase, Acc1, respectively, were sufficient to increase mitochondrial ATP production and quiescence entry. These results suggested that a normal PE synthesis mediated by Ups2-Mdm35 complex attenuates Snf1/AMPK activity, and that Snf1-mediated regulation of carbon metabolisms has great impacts on mitochondrial energy metabolism and quiescence entry. We also found that depletion of Ups2 together with the cell-cycle regulators Whi5 and Whi7, functional orthologs of the Rb1 tumor suppressor, caused a synthetic growth defect in yeast. Similarly, knockdown of PRELID3b, the human homolog of Ups2, decreased the viability of Rb1-deficient breast cancer cells, suggesting that PRELID3b is a potential target for cancer therapy.
Collapse
Affiliation(s)
- Non Miyata
- Department of Chemistry, Faculty of Science, Kyushu University, Fukuoka, Japan
| | - Takanori Ito
- Department of Chemistry, Faculty of Science, Kyushu University, Fukuoka, Japan
| | - Miyu Nakashima
- Department of Chemistry, Faculty of Science, Kyushu University, Fukuoka, Japan
| | - Satoru Fujii
- Department of Chemistry, Faculty of Science, Kyushu University, Fukuoka, Japan
| | - Osamu Kuge
- Department of Chemistry, Faculty of Science, Kyushu University, Fukuoka, Japan
| |
Collapse
|
48
|
Maitiabula G, Tian F, Wang P, Zhang L, Gao X, Wan S, Sun H, Yang J, Zhang Y, Gao T, Xue B, Li C, Li J, Wang X. Liver PP2A-Cα Protects From Parenteral Nutrition-associated Hepatic Steatosis. Cell Mol Gastroenterol Hepatol 2022; 14:669-692. [PMID: 35643235 PMCID: PMC9421584 DOI: 10.1016/j.jcmgh.2022.05.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 05/18/2022] [Accepted: 05/18/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND & AIMS Parenteral nutrition (PN) is a lifesaving therapy for patients with intestinal failure. Hepatic steatosis is a potentially fatal complication of long-term PN, but the involved pathological mechanisms are incompletely unclarified. Herein, we identify the role of protein phosphatase 2A (PP2A) in the pathogenesis of parenteral nutrition-associated hepatic steatosis (PNAHS). METHODS Proteomic/phosphoproteomic analyses of liver samples from patients with PNAHS were applied to identify the mechanism of PNAHS. Total parenteral nutrition (TPN) mice model, in vivo, and in vitro experiments were used to assess the effect of PP2A-Cα on liver fatty acid metabolism. RESULTS Reduced expression of PP2A-Cα (catalytic subunit) enhanced activation of serine/threonine kinase Akt2 and decreased activation of adenosine monophosphate-activated protein kinase (AMPK) were associated with hepatic steatosis in patients with PNAHS. Mice given PN for 14 days developed hepatic steatosis, down-regulation of PP2A-Cα, activation of Akt2, and inhibition of AMPK. Hepatocyte-specific deletion of PP2A-Cα in mice given PN exacerbated Akt2 activation, AMPK inhibition, and hepatic steatosis through an effect on fatty acid degradation, whereas hepatocyte-specific PP2A-Cα overexpression significantly ameliorated hepatic steatosis accompanied with Akt2 suppression and AMPK activation. Additionally, pharmacological activation of Akt2 in mice overexpressing PP2A-Cα led to the aggravation of hepatic steatosis. CONCLUSIONS Our findings demonstrate that hepatic PP2A-Cα serves as a protective factor of PNAHS due to ameliorating hepatic steatosis and improving liver function. Our study provides a strong rationale that PP2A-Cα may be involved in the pathogenesis of PNAHS.
Collapse
Affiliation(s)
- Gulisudumu Maitiabula
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Feng Tian
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Peng Wang
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Li Zhang
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xuejin Gao
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Songlin Wan
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Haifeng Sun
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jianbo Yang
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yupeng Zhang
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Tingting Gao
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Bin Xue
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center of the Medical School of Nanjing University, Nanjing, China,Core Laboratory, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China,Bin Xue, PhD, LongMian Avenue, Nanjing 211166, China. tel: +86-25-87115542
| | - Chaojun Li
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center of the Medical School of Nanjing University, Nanjing, China,Chaojun Li, PhD, Hankou Road, Nanjing, 210093, China. tel: +86-25-83596289.
| | - Jieshou Li
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xinying Wang
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China,Correspondence Address correspondence to: Xinying Wang, MD, PhD, Department of General Surgery, Jinling Hospital, Medical School of Nanjing University. 305 East Zhongshan Road, Nanjing, 210002, China. tel: +86-25-80861429
| |
Collapse
|
49
|
Patel SJ, Liu N, Piaker S, Gulko A, Andrade ML, Heyward FD, Sermersheim T, Edinger N, Srinivasan H, Emont MP, Westcott GP, Luther J, Chung RT, Yan S, Kumari M, Thomas R, Deleye Y, Tchernof A, White PJ, Baselli GA, Meroni M, De Jesus DF, Ahmad R, Kulkarni RN, Valenti L, Tsai L, Rosen ED. Hepatic IRF3 fuels dysglycemia in obesity through direct regulation of Ppp2r1b. Sci Transl Med 2022; 14:eabh3831. [PMID: 35320000 PMCID: PMC9162056 DOI: 10.1126/scitranslmed.abh3831] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Inflammation has profound but poorly understood effects on metabolism, especially in the context of obesity and nonalcoholic fatty liver disease (NAFLD). Here, we report that hepatic interferon regulatory factor 3 (IRF3) is a direct transcriptional regulator of glucose homeostasis through induction of Ppp2r1b, a component of serine/threonine phosphatase PP2A, and subsequent suppression of glucose production. Global ablation of IRF3 in mice on a high-fat diet protected against both steatosis and dysglycemia, whereas hepatocyte-specific loss of IRF3 affects only dysglycemia. Integration of the IRF3-dependent transcriptome and cistrome in mouse hepatocytes identifies Ppp2r1b as a direct IRF3 target responsible for mediating its metabolic actions on glucose homeostasis. IRF3-mediated induction of Ppp2r1b amplified PP2A activity, with subsequent dephosphorylation of AMPKα and AKT. Furthermore, suppression of hepatic Irf3 expression with antisense oligonucleotides reversed obesity-induced insulin resistance and restored glucose homeostasis in obese mice. Obese humans with NAFLD displayed enhanced activation of liver IRF3, with reversion after bariatric surgery. Hepatic PPP2R1B expression correlated with HgbA1C and was elevated in obese humans with impaired fasting glucose. We therefore identify the hepatic IRF3-PPP2R1B axis as a causal link between obesity-induced inflammation and dysglycemia and suggest an approach for limiting the metabolic dysfunction accompanying obesity-associated NAFLD.
Collapse
Affiliation(s)
- Suraj J. Patel
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
- Division of Digestive and Liver Diseases, Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Nan Liu
- Harvard Medical School, Boston, MA 02115, USA
- Cancer and Blood Disorders Center, Dana Farber Cancer Institute and Boston Children’s Hospital, Boston, MA 02215, USA
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China
| | - Sam Piaker
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Anton Gulko
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Maynara L. Andrade
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Frankie D. Heyward
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Tyler Sermersheim
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Nufar Edinger
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Harini Srinivasan
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Margo P. Emont
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Gregory P. Westcott
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Jay Luther
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Raymond T. Chung
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Shuai Yan
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Manju Kumari
- Department of Cardiology, Internal Medicine III, University of Heidelberg, Heidelberg, Germany
| | - Reeby Thomas
- Immunology and Microbiology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Yann Deleye
- Duke Molecular Physiology Institute and Division of Endocrinology, Metabolism and Nutrition, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - André Tchernof
- Institut Universitaire de Cardiologie and Pneumologie de Québec–Université Laval (IUCPQUL), Québec City, Canada
| | - Phillip J. White
- Duke Molecular Physiology Institute and Division of Endocrinology, Metabolism and Nutrition, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Guido A. Baselli
- Department of Pathophysiology and Transplantation, Universita degli Studi di Milano, Milan, Italy
- Precision Medicine, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marica Meroni
- General Medicine and Metabolic Diseases, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Dario F. De Jesus
- Harvard Medical School, Boston, MA 02115, USA
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02215, USA
| | - Rasheed Ahmad
- Immunology and Microbiology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Rohit N. Kulkarni
- Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02215, USA
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, Universita degli Studi di Milano, Milan, Italy
- Precision Medicine, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Linus Tsai
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Evan D. Rosen
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| |
Collapse
|
50
|
Yeon Park S, Cho W, Abd El-Aty A, Hacimuftuoglu A, Hoon Jeong J, Woo Jung T. Valdecoxib attenuates lipid-induced hepatic steatosis through autophagy-mediated suppression of endoplasmic reticulum stress. Biochem Pharmacol 2022; 199:115022. [DOI: 10.1016/j.bcp.2022.115022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 02/09/2023]
|