1
|
Ding Q, Cao W, Ge X, Cao F, Song Q, Jin Y, Sun T, Fang H, Li J, Li S. SMPD3 Inhibition Contributes to Nicotinamide-Ameliorated Hepatic Steatosis in Chronic Alcohol-Fed Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025. [PMID: 40404566 DOI: 10.1021/acs.jafc.5c01450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2025]
Abstract
Alcohol-associated liver disease (ALD) is characterized by the reduction of hepatic nicotinamide adenine dinucleotide (NAD+), which exacerbates hepatic steatosis. The present study was conducted to investigate the protective role of nicotinamide (NAM), a foodborne precursor of NAD+ biosynthesis, in ALD. C57BL/6N mice were employed to establish the ALD model and were administered NAM by gavage. Our results showed that NAM supplementation significantly ameliorated alcohol-induced NAD+ reduction and lipid accumulation in both mice liver and cultured AML-12 hepatocytes and improved lipid metabolism-associated gene disorders. Alcohol-induced liver injury and oxidative stress were also blocked by NAM administration. Further transcriptomics analysis and validation revealed that alcohol-stimulated sphingomyelin phosphodiesterase 3 (SMPD3) was significantly reversed by NAM, along with the reduction of hepatic ceramide levels. Importantly, SMPD3 was upregulated in the livers of ALD patients. Genetically silencing SMPD3 alleviated alcohol-induced lipid accumulation in hepatocytes. ChIP assay identified SMPD3 as a direct downstream target of hypoxia-inducible factor 1 alpha (HIF-1α). Liver-specific Hif1α knockdown reduced the level of hepatic SMPD3 expression in mice. Activation of HIF-1α abolished the prevention of intrahepatic liver lipid deposition by NAM, while SMPD3 knockdown reversed HIF-1α activation-stimulated lipid accumulation, indicating that a HIF-1α-regulated SMPD3 pathway was involved in the beneficial role of NAM. NAM improved liver oxidative stress, while antioxidant MitoQ administration rescued HIF-1α/SMPD3 activation in ALD mice, implying that the antioxidant effect of NAM contributed to its inhibitory role on the HIF-1α/SMPD3 pathway. In conclusion, NAM ameliorates chronic alcohol intake-induced hepatic steatosis by inhibiting SMPD3. This study provides new insights into the mechanistic understanding of ALD and highlights NAM as a therapeutic choice for ALD treatment.
Collapse
Affiliation(s)
- Qinchao Ding
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P. R. China
| | - Wenjing Cao
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P. R. China
| | - Xinxuan Ge
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P. R. China
| | - Feiwei Cao
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P. R. China
| | - Qing Song
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P. R. China
| | - Yue Jin
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P. R. China
| | - Tianchi Sun
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P. R. China
| | - Haoyi Fang
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P. R. China
| | - Jiaxin Li
- Division of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Songtao Li
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P. R. China
| |
Collapse
|
2
|
Dhapola R, Kumari S, Sharma P, Vellingiri B, HariKrishnaReddy D. Advancements in autophagy perturbations in Alzheimer's disease: Molecular aspects and therapeutics. Brain Res 2025; 1851:149494. [PMID: 39922409 DOI: 10.1016/j.brainres.2025.149494] [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: 09/23/2024] [Revised: 12/11/2024] [Accepted: 02/02/2025] [Indexed: 02/10/2025]
Abstract
Emerging evidences suggest that autophagy, a key cellular process responsible for degrading and recycling damaged organelles and proteins, plays a crucial role in maintaining neuronal health. Dysfunctional autophagy has been linked to the pathogenesis of Alzheimer's disease (AD), contributing to the accumulation of misfolded proteins and cellular debris. Molecular mechanisms underlying autophagy dysfunction in AD involve amyloid-beta (Aβ) and tau accumulation, neuroinflammation, mitochondrial dysfunction, oxidative stress and endoplasmic reticulum stress. Disrupted signaling pathways such as TRIB3, Nmnat and BAG3 that regulate key processes like autophagosome initiation, lysosome function, and protein homeostasis also play a crucial role in the pathogenesis. Restoration of autophagy by modulating these molecular and signaling pathways may be an effective therapeutic strategy for AD. Studies have found few drugs targeting autophagy dysregulation in AD. These drugs include metformin that has been found to modulate the expression of TRIB3 for autophagy regulation. Another drug, resveratrol has been reported to augment the activity of Nmnat thus, increases autophagy flux. BACE1 and mTOR inhibitors like arctigenin, nilvadipine and dapagliflozin were also found to restore autophagy. This study elaborates recent advances in signaling and molecular pathways and discusses current and emerging therapeutic interventions targeting autophagy dysfunction in AD.
Collapse
Affiliation(s)
- Rishika Dhapola
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda 151401 Punjab, India
| | - Sneha Kumari
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda 151401 Punjab, India
| | - Prajjwal Sharma
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda 151401 Punjab, India
| | - Balachandar Vellingiri
- Human Cytogenetics and Stem Cell Laboratory, Department of Zoology, School of Basic Sciences, Central University of Punjab 151401 Bathinda, Punjab, India.
| | - Dibbanti HariKrishnaReddy
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda 151401 Punjab, India.
| |
Collapse
|
3
|
Liu Z, Peng H, Liu P, Duan F, Yang Y, Li P, Li Z, Wu J, Chang J, Shang D, Tian Q, Zhang J, Xie Y, Liu Z, An Y. Deciphering significances of autophagy in the development and metabolism of adipose tissue. Exp Cell Res 2025; 446:114478. [PMID: 39978716 DOI: 10.1016/j.yexcr.2025.114478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/17/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025]
Abstract
The mechanisms of adipose tissue activation and inactivation have been a hot topic of research in the last decade, from which countermeasures have been attempted to be found against obesity as well as other lipid metabolism-related diseases, such as type 2 diabetes mellitus and non-alcoholic fatty liver disease. Autophagy has been shown to be closely related to the regulation of adipocyte activity, which is involved in the whole process including white adipocyte differentiation/maturation and brown or beige adipocyte generation/activation. Dysregulation of autophagy in adipose tissue has been demonstrated to be associated with obesity. On this basis, we summarize the pathways and mechanisms of autophagy involved in the regulation of lipid metabolism and present a review of its pathophysiological roles in lipid metabolism-related diseases, in the hope of providing ideas for the treatment of these diseases.
Collapse
Affiliation(s)
- Zitao Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Haoyuan Peng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Pengfei Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Feiyi Duan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Yutian Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Pengkun Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Zhihao Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Jiaoyan Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Jiayi Chang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Dandan Shang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Qiwen Tian
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Jiawei Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Yucheng Xie
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Zhenzhen Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Yang An
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China; Henan Provincial Research Center of Engineering Technology for Nuclear Protein Medical Detection, Zhengzhou Health College, Zhengzhou, 450064, China.
| |
Collapse
|
4
|
Zhang R, Yan Z, Zhong H, Luo R, Liu W, Xiong S, Liu Q, Liu M. Gut microbial metabolites in MASLD: Implications of mitochondrial dysfunction in the pathogenesis and treatment. Hepatol Commun 2024; 8:e0484. [PMID: 38967596 PMCID: PMC11227362 DOI: 10.1097/hc9.0000000000000484] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/09/2024] [Indexed: 07/06/2024] Open
Abstract
With an increasing prevalence, metabolic dysfunction-associated steatotic liver disease (MASLD) has become a major global health problem. MASLD is well-known as a multifactorial disease. Mitochondrial dysfunction and alterations in the gut bacteria are 2 vital events in MASLD. Recent studies have highlighted the cross-talk between microbiota and mitochondria, and mitochondria are recognized as pivotal targets of the gut microbiota to modulate the host's physiological state. Mitochondrial dysfunction plays a vital role in MASLD and is associated with multiple pathological changes, including hepatocyte steatosis, oxidative stress, inflammation, and fibrosis. Metabolites are crucial mediators of the gut microbiota that influence extraintestinal organs. Additionally, regulation of the composition of gut bacteria may serve as a promising therapeutic strategy for MASLD. This study reviewed the potential roles of several common metabolites in MASLD, emphasizing their impact on mitochondrial function. Finally, we discuss the current treatments for MASLD, including probiotics, prebiotics, antibiotics, and fecal microbiota transplantation. These methods concentrate on restoring the gut microbiota to promote host health.
Collapse
Affiliation(s)
- Ruhan Zhang
- College of Acupuncture, Tuina, and Rehabilitation, Hunan University of Chinese Medicine, Hunan, China
| | - Zhaobo Yan
- College of Acupuncture, Tuina, and Rehabilitation, Hunan University of Chinese Medicine, Hunan, China
| | - Huan Zhong
- College of Acupuncture, Tuina, and Rehabilitation, Hunan University of Chinese Medicine, Hunan, China
| | - Rong Luo
- Department of Acupuncture and Massage Rehabilitation, The First Affiliated Hospital of Hunan University of Chinese Medicine, Hunan, China
| | - Weiai Liu
- Department of Acupuncture and Massage Rehabilitation, The Second Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Hunan, China
| | - Shulin Xiong
- Department of Preventive Center, The Second Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Hunan, China
| | - Qianyan Liu
- College of Acupuncture, Tuina, and Rehabilitation, Hunan University of Chinese Medicine, Hunan, China
| | - Mi Liu
- College of Acupuncture, Tuina, and Rehabilitation, Hunan University of Chinese Medicine, Hunan, China
| |
Collapse
|
5
|
Tian C, Huang R, Xiang M. SIRT1: Harnessing multiple pathways to hinder NAFLD. Pharmacol Res 2024; 203:107155. [PMID: 38527697 DOI: 10.1016/j.phrs.2024.107155] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/04/2024] [Accepted: 03/21/2024] [Indexed: 03/27/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) encompasses hepatic steatosis, non-alcoholic steatohepatitis (NASH), fibrosis, cirrhosis, and hepatocellular carcinoma. It is the primary cause of chronic liver disorders, with a high prevalence but no approved treatment. Therefore, it is indispensable to find a trustworthy therapy for NAFLD. Recently, mounting evidence illustrates that Sirtuin 1 (SIRT1) is strongly associated with NAFLD. SIRT1 activation or overexpression attenuate NAFLD, while SIRT1 deficiency aggravates NAFLD. Besides, an array of therapeutic agents, including natural compounds, synthetic compounds, traditional Chinese medicine formula, and stem cell transplantation, alleviates NALFD via SIRT1 activation or upregulation. Mechanically, SIRT1 alleviates NAFLD by reestablishing autophagy, enhancing mitochondrial function, suppressing oxidative stress, and coordinating lipid metabolism, as well as reducing hepatocyte apoptosis and inflammation. In this review, we introduced the structure and function of SIRT1 briefly, and summarized the effect of SIRT1 on NAFLD and its mechanism, along with the application of SIRT1 agonists in treating NAFLD.
Collapse
Affiliation(s)
- Cheng Tian
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Rongrong Huang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ming Xiang
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
6
|
Dashti Z, Yousefi Z, Kiani P, Taghizadeh M, Maleki MH, Borji M, Vakili O, Shafiee SM. Autophagy and the unfolded protein response shape the non-alcoholic fatty liver landscape: decoding the labyrinth. Metabolism 2024; 154:155811. [PMID: 38309690 DOI: 10.1016/j.metabol.2024.155811] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/23/2024] [Accepted: 01/28/2024] [Indexed: 02/05/2024]
Abstract
The incidence of nonalcoholic fatty liver disease (NAFLD) is on the rise, mirroring a global surge in diabetes and metabolic syndrome, as its major leading causes. NAFLD represents a spectrum of liver disorders, ranging from nonalcoholic fatty liver (NAFL) to nonalcoholic steatohepatitis (NASH), which can potentially progress to cirrhosis and hepatocellular carcinoma (HCC). Mechanistically, we know the unfolded protein response (UPR) as a protective cellular mechanism, being triggered under circumstances of endoplasmic reticulum (ER) stress. The hepatic UPR is turned on in a broad spectrum of liver diseases, including NAFLD. Recent data also defines molecular mechanisms that may underlie the existing correlation between UPR activation and NAFLD. More interestingly, subsequent studies have demonstrated an additional mechanism, i.e. autophagy, to be involved in hepatic steatosis, and thus NAFLD pathogenesis, principally by regulating the insulin sensitivity, hepatocellular injury, innate immunity, fibrosis, and carcinogenesis. All these findings suggest possible mechanistic roles for autophagy in the progression of NAFLD and its complications. Both UPR and autophagy are dynamic and interconnected fluxes that act as protective responses to minimize the harmful effects of hepatic lipid accumulation, as well as the ER stress during NAFLD. The functions of UPR and autophagy in the liver, together with findings of decreased hepatic autophagy in correlation with conditions that predispose to NAFLD, such as obesity and aging, suggest that autophagy and UPR, alone or combined, may be novel therapeutic targets against the disease. In this review, we discuss the current evidence on the interplay between autophagy and the UPR in connection to the NAFLD pathogenesis.
Collapse
Affiliation(s)
- Zahra Dashti
- Department of Genetics, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Zeynab Yousefi
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Pouria Kiani
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Motahareh Taghizadeh
- Department of Clinical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Hasan Maleki
- Department of Clinical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Borji
- Department of Clinical Biochemistry, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Omid Vakili
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran; Autophagy Research Center, Department of Clinical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Sayed Mohammad Shafiee
- Autophagy Research Center, Department of Clinical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
7
|
Zhang D, Ma Y, Liu J, Wang D, Geng Z, Wen D, Chen H, Wang H, Li L, Zhu X, Wang X, Huang M, Zou C, Chen Y, Ma L. Fenofibrate improves hepatic steatosis, insulin resistance, and shapes the gut microbiome via TFEB-autophagy in NAFLD mice. Eur J Pharmacol 2023; 960:176159. [PMID: 37898287 DOI: 10.1016/j.ejphar.2023.176159] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 10/23/2023] [Accepted: 10/24/2023] [Indexed: 10/30/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a major liver disease subtype worldwide, is commonly associated with insulin resistance and obesity. NAFLD is characterized by an excessive hepatic lipid accumulation, as well as hepatic steatosis. Fenofibrate is a peroxisome proliferator-activated receptor α agonist widely used in clinical therapy to effectively ameliorate the development of NAFLD, but its mechanism of action is incompletely understood. Here, we found that fenofibrate dramatically modulate the gut microbiota composition of high-fat diet (HFD)-induced NAFLD mouse model, and the change of gut microbiota composition is dependent on TFEB-autophagy axis. Furthermore, we also found that fenofibrate improved hepatic steatosis, and increased the activation of TFEB, which severed as a regulator of autophagy, thus, the protective effects of fenofibrate against NAFLD are depended on TFEB-autophagy axis. Our study demonstrates the host gene may influence the gut microbiota and highlights the role of TFEB and autophagy in the protective effect of NAFLD. This work expands our understanding of the regulatory interactions between the host and gut microbiota and provides novel strategies for alleviating obesity.
Collapse
Affiliation(s)
- Dan Zhang
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, 650032, PR China
| | - Yicheng Ma
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, 650091, PR China
| | - Jianjun Liu
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Biomedical Engineering, Kunming Medical University, Kunming, 650500, PR China
| | - Da Wang
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, 650032, PR China
| | - Zuotao Geng
- Department of Pediatrics, Women and Children's Hospital of Lijiang, Lijiang, 674100, PR China
| | - Daiyan Wen
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, 650032, PR China
| | - Hang Chen
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, 650032, PR China
| | - Hui Wang
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, 650032, PR China
| | - Lanyi Li
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, 650032, PR China
| | - Xiaotong Zhu
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, 650032, PR China
| | - Xuemin Wang
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, 650032, PR China
| | - Minshan Huang
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, 650032, PR China
| | - Chenggang Zou
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, 650091, PR China.
| | - Yuanli Chen
- Faculty of Basic Medicine, Kunming Medical University, Kunming, 650500, PR China.
| | - Lanqing Ma
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, 650032, PR China.
| |
Collapse
|
8
|
Qiu J, Chen L, Zhang L, Xu F, Zhang C, Ren G, Chang K, He G, Du Z, Le Y, Yu Z, Li S, Liu Q, Dou X. Xie Zhuo Tiao Zhi formula modulates intestinal microbiota and liver purine metabolism to suppress hepatic steatosis and pyroptosis in NAFLD therapy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 121:155111. [PMID: 37804819 DOI: 10.1016/j.phymed.2023.155111] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/05/2023] [Accepted: 09/19/2023] [Indexed: 10/09/2023]
Abstract
BACKGROUND Current evidence indicates a rising global prevalence of Non-Alcoholic Fatty Liver Disease (NAFLD), which is closely associated to conditions such as obesity, dyslipidemia, insulin resistance, and metabolic syndrome. The relationship between the gut microbiome and metabolites in NAFLD is gaining attention understanding the pathogenesis and progression of dysregulated lipid metabolism and inflammation. The Xie Zhuo Tiao Zhi (XZTZ) decoction has been employed in clinical practice for alleviating hyperlipidemia and symptoms related to metabolic disorders. However, the pharmacological mechanisms underlying the effects of XZTZ remain to be elucidated. PURPOSE The objective of this study was to examine the pharmacological mechanisms underlying the hypolipidemic and anti-inflammatory effects of XZTZ decoction in a mouse model of NAFLD, as well as the effects of supplementing exogenous metabolites on PO induced cell damage and lipid accumulation in cultured hepatocytes. METHODS A high-fat diet (HFD) mouse model was established to examine the effects of XZTZ through oral gavage. The general condition of mice and the protective effect of XZTZ on liver injury were evaluated using histological and biochemical methods. Hematoxylin and eosin staining (H&E) staining and oil red O staining were performed to assess inflammatory and lipid accumulation detection, and cytokine levels were quantitatively analyzed. Additionally, the study included full-length 16S rRNA sequencing, liver transcriptome analysis, and non-targeted metabolomics analysis to investigate the relationship among intestinal microbiome, liver metabolic function, and XZTZ decoction. RESULTS XZTZ had a significant impact on the microbial community structure in NAFLD mice. Notably, the abundance of Ileibacterium valens, which was significantly enriched by XZTZ, exhibited a negative correlation with liver injury biomarkers such as, alanine transaminase (ALT) and aspartate transaminase (AST) activity. Moreover, treatment with XZTZ led to a significant enrichment of the purine metabolism pathway in liver tissue metabolites, with inosine, a purine metabolite, showing a significant positive correlation with the abundance of I. valens. XZTZ and inosine also significantly enhanced fatty acid β-oxidation, which led to a reduction in the expression of pro-inflammatory cytokines and the inhibition of liver pyroptosis. These effects contributed to the mitigation of liver injury and hepatocyte damage, both in vivo and vitro. Furthermore, the utilization of HPLC fingerprints and UPLC-Q-TOF-MS elucidated the principal constituents within the XZTZ decoction, including naringin, neohesperidin, atractylenolide III, 23-o-Acetylalisol B, pachymic acid, and ursolic acid which are likely responsible for its therapeutic efficacy. Further investigations are imperative to fully uncover and validate the pharmacodynamic mechanisms underlying these observations. CONCLUSION The administration of XZTZ decoction demonstrates a protective effect on the livers of NAFLD mice by inhibiting lipid accumulation and reducing hepatocyte inflammatory damage. This protective effect is mediated by the upregulation of I.valens abundance in the intestine, highlighting the importance of the gut-liver axis. Furthermore, the presesnce of inosine, adenosine, and their derivatives are important in promoting the protective effects of XZTZ. Furthermore, the in vitro approaching, we provide hitherto undocumented evidence indicating that the inosine significantly improves lipid accumulation, inflammatory damage, and pyroptosis in AML12 cells incubated with free fatty acids.
Collapse
Affiliation(s)
- Jiannan Qiu
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Lin Chen
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Ling Zhang
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Fangying Xu
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Congcong Zhang
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Guilin Ren
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Kaixin Chang
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Guonong He
- Ningbo Traditional Chinese Medicine Hospital, Ningbo, Zhejiang, PR China
| | - Zhongyan Du
- School of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Yifei Le
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Zhiling Yu
- Consun Chinese Medicines Research Centre for Renal Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Songtao Li
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Qingsheng Liu
- Hangzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang, PR China.
| | - Xiaobing Dou
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China.
| |
Collapse
|
9
|
Afshari H, Noori S, Zarghi A. A novel combination of metformin and resveratrol alleviates hepatic steatosis by activating autophagy through the cAMP/AMPK/SIRT1 signaling pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:3135-3148. [PMID: 37209153 DOI: 10.1007/s00210-023-02520-7] [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: 02/08/2023] [Accepted: 05/03/2023] [Indexed: 05/22/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a prevalent liver disorder that is associated with the accumulation of triglycerides (TG) in hepatocytes. Resveratrol (RSV), as a natural product, and metformin have been reported to have potential lipid-lowering effects for the treatment of NAFLD via autophagy, but the combined effects of both have not yet been studied. The current study aimed to investigate the role of autophagy in the lipid-lowering effects of RSV, alone and in combination with metformin, on the hepatic steatosis model of HepG2 cells and elucidate the mechanism of action. Triglyceride measurement and real-time PCR showed that RSV-metformin reduced lipid accumulation and the expression of lipogenic genes in palmitic acid (PA)-induced HepG2 cells. Additionally, the LDH release assay indicated that this combination protected HepG2 cells against PA-induced cell death through autophagy. The western blotting analysis revealed that RSV-metformin induced autophagy by reducing the expression of p62 and increasing LC3-I and LC3-II proteins. This combination also enhanced cAMP, phosphorylated AMP-activated protein kinase (p-AMPK), and Beclin-1 levels in HepG2 cells. Furthermore, SIRT1 inhibitor treatment inhibited autophagy induced by RSV-metformin, which indicated the autophagy induction is SIRT1-dependent. This study demonstrated for the first time that RSV-metformin reduced hepatic steatosis by triggering autophagy via the cAMP/AMPK/SIRT1 signaling pathway.
Collapse
Affiliation(s)
- Havva Afshari
- Department of Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shokoofe Noori
- Department of Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Afshin Zarghi
- Department of Pharmaceutical Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
ZHANG YU, ZHOU XI, ZHANG CHUNYAN, LAI DENGNI, LIU DONGBO, WU YANYANG. Vitamin B3 inhibits apoptosis and promotes autophagy of islet β cells under high glucose stress. BIOCELL 2023. [DOI: 10.32604/biocell.2023.026429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2023]
|
11
|
Hu G, Ling C, Chi L, Thind MK, Furse S, Koulman A, Swann JR, Lee D, Calon MM, Bourdon C, Versloot CJ, Bakker BM, Gonzales GB, Kim PK, Bandsma RHJ. The role of the tryptophan-NAD + pathway in a mouse model of severe malnutrition induced liver dysfunction. Nat Commun 2022; 13:7576. [PMID: 36481684 PMCID: PMC9732354 DOI: 10.1038/s41467-022-35317-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 11/29/2022] [Indexed: 12/13/2022] Open
Abstract
Mortality in children with severe malnutrition is strongly related to signs of metabolic dysfunction, such as hypoglycemia. Lower circulating tryptophan levels in children with severe malnutrition suggest a possible disturbance in the tryptophan-nicotinamide adenine dinucleotide (TRP-NAD+) pathway and subsequently in NAD+ dependent metabolism regulator sirtuin1 (SIRT1). Here we show that severe malnutrition in weanling mice, induced by 2-weeks of low protein diet feeding from weaning, leads to an impaired TRP-NAD+ pathway with decreased NAD+ levels and affects hepatic mitochondrial turnover and function. We demonstrate that stimulating the TRP-NAD+ pathway with NAD+ precursors improves hepatic mitochondrial and overall metabolic function through SIRT1 modulation. Activating SIRT1 is sufficient to induce improvement in metabolic functions. Our findings indicate that modulating the TRP-NAD+ pathway can improve liver metabolic function in a mouse model of severe malnutrition. These results could lead to the development of new interventions for children with severe malnutrition.
Collapse
Affiliation(s)
- Guanlan Hu
- grid.17063.330000 0001 2157 2938Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, M5G 1A8 Toronto, Canada ,grid.42327.300000 0004 0473 9646Translational Medicine Program, The Hospital for Sick Children, M5G 0A4 Toronto, Canada
| | - Catriona Ling
- grid.17063.330000 0001 2157 2938Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, M5G 1A8 Toronto, Canada ,grid.42327.300000 0004 0473 9646Translational Medicine Program, The Hospital for Sick Children, M5G 0A4 Toronto, Canada
| | - Lijun Chi
- grid.42327.300000 0004 0473 9646Translational Medicine Program, The Hospital for Sick Children, M5G 0A4 Toronto, Canada
| | - Mehakpreet K. Thind
- grid.17063.330000 0001 2157 2938Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, M5G 1A8 Toronto, Canada ,grid.42327.300000 0004 0473 9646Translational Medicine Program, The Hospital for Sick Children, M5G 0A4 Toronto, Canada
| | - Samuel Furse
- grid.5335.00000000121885934Core Metabolomics and Lipidomics Laboratory, Wellcome Trust-Metabolic Research Laboratories, Institute of Metabolic Sciences, University of Cambridge, CB2 0QQ Cambridge, UK ,grid.4903.e0000 0001 2097 4353Biological Chemistry Group, Royal Botanic Gardens, Kew, Kew Green, TW9 3AE Richmond, UK
| | - Albert Koulman
- grid.5335.00000000121885934Core Metabolomics and Lipidomics Laboratory, Wellcome Trust-Metabolic Research Laboratories, Institute of Metabolic Sciences, University of Cambridge, CB2 0QQ Cambridge, UK
| | - Jonathan R. Swann
- grid.5491.90000 0004 1936 9297School of Human Development and Health, Faculty of Medicine, University of Southampton, SO16 6YD Southampton, UK ,grid.7445.20000 0001 2113 8111Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, SW7 2AZ London, UK
| | - Dorothy Lee
- grid.42327.300000 0004 0473 9646Translational Medicine Program, The Hospital for Sick Children, M5G 0A4 Toronto, Canada
| | - Marjolein M. Calon
- grid.42327.300000 0004 0473 9646Translational Medicine Program, The Hospital for Sick Children, M5G 0A4 Toronto, Canada
| | - Celine Bourdon
- grid.42327.300000 0004 0473 9646Translational Medicine Program, The Hospital for Sick Children, M5G 0A4 Toronto, Canada ,grid.511677.3The Childhood Acute Illness & Nutrition Network (CHAIN), Nairobi, Kenya
| | - Christian J. Versloot
- grid.4494.d0000 0000 9558 4598Laboratory of Pediatrics, Center for Liver, Digestive, and Metabolic Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Barbara M. Bakker
- grid.4494.d0000 0000 9558 4598Laboratory of Pediatrics, Center for Liver, Digestive, and Metabolic Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Gerard Bryan Gonzales
- grid.42327.300000 0004 0473 9646Translational Medicine Program, The Hospital for Sick Children, M5G 0A4 Toronto, Canada ,grid.4818.50000 0001 0791 5666Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Peter K. Kim
- grid.17063.330000 0001 2157 2938Department of Biochemistry, University of Toronto, M5S 1A8 Toronto, Canada ,grid.42327.300000 0004 0473 9646Cell Biology Program, The Hospital for Sick Children, M5G 0A4 Toronto, Canada
| | - Robert H. J. Bandsma
- grid.17063.330000 0001 2157 2938Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, M5G 1A8 Toronto, Canada ,grid.42327.300000 0004 0473 9646Translational Medicine Program, The Hospital for Sick Children, M5G 0A4 Toronto, Canada ,grid.511677.3The Childhood Acute Illness & Nutrition Network (CHAIN), Nairobi, Kenya ,grid.4494.d0000 0000 9558 4598Laboratory of Pediatrics, Center for Liver, Digestive, and Metabolic Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands ,grid.42327.300000 0004 0473 9646Division of Gastroenterology, Hepatology, and Nutrition, The Hospital for Sick Children, M5G 0A4 Toronto, Canada
| |
Collapse
|
12
|
Hofer SJ, Kroemer G, Kepp O. Autophagy-inducing nutritional interventions in experimental and clinical oncology. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 373:125-158. [PMID: 36283765 DOI: 10.1016/bs.ircmb.2022.08.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Numerous pro-autophagic dietary interventions are being investigated for their potential cancer-preventive or therapeutic effects. This applies to different fasting regimens, methionine restriction and ketogenic diets. In addition, the supplementation of specific micronutrients such as nicotinamide (vitamin B3) or spermidine induces autophagy. In humans, leanness, plant-based diets (that may lead to partial methionine restriction) and high dietary uptake of spermidine are associated with a low incidence of cancers. Moreover, clinical trials have demonstrated the capacity of nicotinamide to prevent non-melanoma skin carcinogenesis. Multiple interventional trials are evaluating the capacity of autophagy-inducing regimens to improve the outcome of chemotherapy and immunotherapy. Here, we discuss the mechanistic underpinnings of autophagy induction by nutritional interventions, as well as the mechanisms through which autophagy induction in malignant or immune cells improves anticancer immunosurveillance.
Collapse
Affiliation(s)
- Sebastian J Hofer
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France; Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Paris, France; Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France; Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Paris, France; Institut du Cancer Paris Carpem, Department of Biology, APHP, Hôpital Européen Georges Pompidou, Paris, France.
| | - Oliver Kepp
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France; Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Paris, France.
| |
Collapse
|
13
|
mTOR: A Potential New Target in Nonalcoholic Fatty Liver Disease. Int J Mol Sci 2022; 23:ijms23169196. [PMID: 36012464 PMCID: PMC9409235 DOI: 10.3390/ijms23169196] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 11/17/2022] Open
Abstract
The global prevalence of nonalcoholic fatty liver disease (NAFLD) continues to rise, yet effective treatments are lacking due to the complex pathogenesis of this disease. Although recent research has provided evidence for the “multiple strikes” theory, the classic “two strikes” theory has not been overturned. Therefore, there is a crucial need to identify multiple targets in NAFLD pathogenesis for the development of diagnostic markers and targeted therapeutics. Since its discovery, the mechanistic target of rapamycin (mTOR) has been recognized as the central node of a network that regulates cell growth and development and is closely related to liver lipid metabolism and other processes. This paper will explore the mechanisms by which mTOR regulates lipid metabolism (SREBPs), insulin resistance (Foxo1, Lipin1), oxidative stress (PIG3, p53, JNK), intestinal microbiota (TLRs), autophagy, inflammation, genetic polymorphisms, and epigenetics in NAFLD. The specific influence of mTOR on NAFLD was hypothesized to be divided into micro regulation (the mechanism of mTOR’s influence on NAFLD factors) and macro mediation (the relationship between various influencing factors) to summarize the influence of mTOR on the developmental process of NAFLD, and prove the importance of mTOR as an influencing factor of NAFLD regarding multiple aspects. The effects of crosstalk between mTOR and its upstream regulators, Notch, Hedgehog, and Hippo, on the occurrence and development of NAFLD-associated hepatocellular carcinoma are also summarized. This analysis will hopefully support the development of diagnostic markers and new therapeutic targets in NAFLD.
Collapse
|
14
|
Fan Y, Dong W, Wang Y, Zhu S, Chai R, Xu Z, Zhang X, Yan Y, Yang L, Bian Y. Glycyrrhetinic acid regulates impaired macrophage autophagic flux in the treatment of non-alcoholic fatty liver disease. Front Immunol 2022; 13:959495. [PMID: 35967372 PMCID: PMC9365971 DOI: 10.3389/fimmu.2022.959495] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 07/01/2022] [Indexed: 11/13/2022] Open
Abstract
Macrophages are involved in hepatocyte steatosis and necroinflammation and play an important role in the pathogenesis of non-alcoholic fatty liver disease (NAFLD). Impaired autophagy function (decreased autophagy or blocked autophagic flow) leads to cell damage and death and promotes NAFLD progression. The experimental and clinical research of glycyrrhetinic acid (GA) in the treatment of NAFLD has gradually attracted attention with clear pharmacological activities such as immune regulation, antiviral, antitumor, antioxidant, liver protection, and anti-inflammatory. However, the effects of GA on the STAT3-HIF-1α pathway and autophagy in macrophages are still unclear, and its mechanism of action in the treatment of NAFLD remains to be further elucidated. We constructed a NAFLD mouse model through a high-fat and high-sugar diet to investigate the therapeutic effects of GA. The results showed that GA reduced weight, improved the pathological changes and hepatic lipid deposition of liver, and abnormally elevated the levels of serum biochemical (AST, ALT, TG, T-CHO, LDL-C, and HDL-C) and inflammatory indexes (IL-1β, IL-4, IL-6, MCP-1, and TNF-α) in NAFLD mice. Further examination revealed that GA ameliorates excessive hepatic macrophage infiltration and hepatocyte apoptosis. The results of the cell experiments further elaborated that GA modulated the PA-induced macrophage STAT3-HIF-1α pathway and ameliorated impaired autophagic flux (blockade of autophagosome–lysosome fusion) and overactivation of inflammation. Excessive hepatocyte apoptosis caused by the uncontrolled release of inflammatory cytokines was also suppressed by GA.ConclusionThis study demonstrated that GA could regulate the STAT3-HIF-1α pathway of macrophages, ameliorate the impaired autophagy flux, and reduce the excessive production of inflammatory cytokines to improve the excessive apoptosis of liver cells, thus playing a therapeutic role on NAFLD.
Collapse
Affiliation(s)
- Yadong Fan
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wenjin Dong
- Department of Science and Education, Tianjin Union Medical Center, Tianjin, China
| | - Ying Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shan Zhu
- State Key Laboratory of Component Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Rundong Chai
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhe Xu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaoyu Zhang
- The Reproductive Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yiqi Yan
- State Key Laboratory of Component Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Long Yang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Research Center for Infectious Diseases, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- *Correspondence: Long Yang, ; Yuhong bian,
| | - Yuhong Bian
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- *Correspondence: Long Yang, ; Yuhong bian,
| |
Collapse
|
15
|
Luo C, Yang C, Wang X, Chen Y, Liu X, Deng H. Nicotinamide reprograms adipose cellular metabolism and increases mitochondrial biogenesis to ameliorate obesity. J Nutr Biochem 2022; 107:109056. [DOI: 10.1016/j.jnutbio.2022.109056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 03/22/2022] [Accepted: 04/06/2022] [Indexed: 11/29/2022]
|
16
|
Cimifugin Ameliorates Lipotoxicity-Induced Hepatocyte Damage and Steatosis through TLR4/p38 MAPK- and SIRT1-Involved Pathways. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4557532. [PMID: 35355867 PMCID: PMC8958062 DOI: 10.1155/2022/4557532] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/24/2022] [Accepted: 03/03/2022] [Indexed: 12/21/2022]
Abstract
Objective. Hepatic metabolic disorder induced by lipotoxicity plays a detrimental role in metabolic fatty liver disease pathogenesis. Cimifugin (Cim), a coumarin derivative extracted from the root of Saposhnikovia divaricata, possesses multiple biological properties against inflammation, allergy, and oxidative stress. However, limited study has addressed the hepatoprotective role of Cim. Here, we investigate the protective effect of Cim against lipotoxicity-induced cytotoxicity and steatosis in hepatocytes and clarify its potential mechanisms. Methods. AML-12, a nontransformed mouse hepatocyte cell line, was employed in this study. The cells were incubated with palmitate or oleate to imitate hepatotoxicity or steatosis model, respectively. Results. Cim significantly reversed palmitate-induced hepatocellular injury in a dose-dependent manner, accompanied by improvements in oxidative stress and mitochondrial damage. Cim pretreatment reversed palmitate-stimulated TLR4/p38 MAPK activation and SIRT1 reduction without affecting JNK, ERK1/2, and AMPK pathways. The hepatoprotective effects of Cim were abolished either through activating TLR4/p38 by their pharmacological agonists or genetical silencing SIRT1 via special siRNA, indicating a mechanistic involvement. Moreover, Cim treatment improved oleate-induced hepatocellular lipid accumulation, which could be blocked by either TLR4 stimulation or SIRT1 knockdown. We observed that SIRT1 was a potential target of TLR4 in palmitate-treated hepatocytes, since TLR4 agonist LPS aggravated, whereas TLR4 antagonist CLI-095 alleviated palmitate-decreased SIRT1 expression. SIRT1 knockdown did not affect palmitate-induced TLR4. In addition, TLR4 activation by LPS significantly abolished Cim-protected SIRT1 reduction induced by palmitate. These results collaboratively indicated that TLR4-regulated SIRT1 pathways was mechanistically involved in the protective effects of Cim against lipotoxicity. Conclusion. In brief, we demonstrate the protective effects of Cim against lipotoxicity-induced cell death and steatosis in hepatocytes. TLR4-regulated p38 MAPK and SIRT1 pathways are involved in Cim-protected hepatic lipotoxicity. Cim is a potential candidate for improving hepatic metabolic disorders mediated by lipotoxicity.
Collapse
|
17
|
El-Kady RR, Ali AK, El Wakeel LM, Sabri NA, Shawki MA. Nicotinamide supplementation in diabetic nonalcoholic fatty liver disease patients: randomized controlled trial. Ther Adv Chronic Dis 2022; 13:20406223221077958. [PMID: 35222903 PMCID: PMC8874180 DOI: 10.1177/20406223221077958] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 01/13/2022] [Indexed: 12/16/2022] Open
Abstract
Background: Nicotinamide has been reported to protect against liver steatosis and metabolic imbalances in nonalcoholic fatty liver disease (NAFLD) in animal models. Objectives: The objective was to investigate the efficacy and safety of nicotinamide supplementation in diabetic NAFLD patients. Design: This is a prospective randomized controlled open label study. Methods: Seventy diabetic NAFLD patients were randomly assigned either to the nicotinamide group (n = 35) who received nicotinamide 1000 mg once daily for 12 weeks in addition to their antidiabetic therapy or the control group (n = 35) who received their antidiabetic therapy only. The primary outcome was improvement in steatosis score, while secondary outcomes included assessment of liver stiffness, liver enzymes, lipid profile, insulin resistance, serum malondialdehyde, serum adiponectin, and patients’ quality of life (QOL). Results: Only 61 patients completed the study; 31 in the nicotinamide group and 30 in the control group. Comparisons between groups and within groups revealed nonsignificant changes in steatosis and fibrosis scores. However, significant reduction was observed in liver enzymes with a median decrease in alanine transaminase of 26.6% versus 0.74% in nicotinamide and control groups, respectively. After 12 weeks of treatment, the nicotinamide group showed significantly lower levels of low-density lipoprotein cholesterol (p value = 0.004), total cholesterol (p value = 0.006), and insulin resistance marker (p value = 0.005) compared with control. Serum triglycerides, malondialdehyde, and adiponectin levels were all comparable between the two groups. Regarding QOL, a significant improvement was detected in the total scores and the activity and fatigue domains scores. Conclusion: Nicotinamide at a dose of 1000 mg daily was tolerable, improved metabolic abnormalities and QOL of diabetic NAFLD patients with no effect on liver fibrosis or steatosis. Trial Registration: The study was registered at clinicaltrials.gov and given the ID number: ‘NCT03850886’. https://clinicaltrials.gov/ct2/show/NCT03850886.
Collapse
Affiliation(s)
- Rasha R. El-Kady
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Amani K. Ali
- Department of Internal Medicine, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Lamia M. El Wakeel
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Nagwa A. Sabri
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - May A. Shawki
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| |
Collapse
|
18
|
Yang HY, Chen JY, Huo YN, Yu PL, Lin PZ, Hsu SC, Huang SM, Tsai CS, Lin CY. The Role of Sirtuin 1 in Palmitic Acid-Induced Endoplasmic Reticulum Stress in Cardiac Myoblasts. Life (Basel) 2022; 12:life12020182. [PMID: 35207470 PMCID: PMC8878829 DOI: 10.3390/life12020182] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/07/2022] [Accepted: 01/24/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Lipotoxicity causes endoplasmic reticulum (ER) stress, leading to cell apoptosis. Sirtuin 1 (Sirt1) regulates gene transcription and cellular metabolism. In this study, we investigated the role of Sirt1 in palmitate-induced ER stress. Methods: Both H9c2 myoblasts and heart-specific Sirt1 knockout mice fed a palmitate-enriched high-fat diet were used. Results: The high-fat diet induced C/EBP homologous protein (CHOP) and activating transcription factor 4 (ATF4) expression in both Sirt1 knockout mice and controls. The Sirt1 knockout mice showed higher CHOP and ATF4 expression compared to those in the control. Palmitic acid (PA) induced ATF4 and CHOP expression in H9c2 cells. PA-treated H9c2 cells showed decreased cytosolic NAD+/NADH alongside reduced Sirt1′s activity. The H9c2 cells showed increased ATF4 and CHOP expression when transfected with plasmid encoding dominant negative mutant Sirt1. Sirt1 activator SRT1720 did not affect CHOP and ATF4 expression. Although SRT1720 enhanced the nuclear translocation of ATF4, the extent of the binding of ATF4 to the CHOP promoter did not increase in PA treated-H9c2 cells. Conclusion: PA-induced ER stress is mediated through the upregulation of ATF4 and CHOP. Cytosolic NAD+ concentration is diminished by PA-induced ER stress, leading to decreased Sirt1 activity. The Sirt1 activator SRT1720 promotes the nuclear translocation of ATF4 in PA-treated H9c2 cells.
Collapse
Affiliation(s)
- Hsiang-Yu Yang
- Division of Cardiovascular Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan;
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan
- Department of Biochemistry, National Defense Medical Center, Taipei 114, Taiwan; (J.-Y.C.); (P.-L.Y.); (P.-Z.L.); (S.-M.H.)
| | - Jhao-Ying Chen
- Department of Biochemistry, National Defense Medical Center, Taipei 114, Taiwan; (J.-Y.C.); (P.-L.Y.); (P.-Z.L.); (S.-M.H.)
| | - Yen-Nien Huo
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 114, Taiwan;
| | - Pei-Ling Yu
- Department of Biochemistry, National Defense Medical Center, Taipei 114, Taiwan; (J.-Y.C.); (P.-L.Y.); (P.-Z.L.); (S.-M.H.)
| | - Pei-Zhen Lin
- Department of Biochemistry, National Defense Medical Center, Taipei 114, Taiwan; (J.-Y.C.); (P.-L.Y.); (P.-Z.L.); (S.-M.H.)
| | | | - Shih-Ming Huang
- Department of Biochemistry, National Defense Medical Center, Taipei 114, Taiwan; (J.-Y.C.); (P.-L.Y.); (P.-Z.L.); (S.-M.H.)
| | - Chien-Sung Tsai
- Division of Cardiovascular Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan;
- Department and Graduate Institute of Pharmacology, National Defense Medical Center, Taipei 114, Taiwan
- Correspondence: (C.-S.T.); (C.-Y.L.); Tel.: +886-2-8792-7212 (C.-Y.L.); Fax: +886-2-8792-7376 (C.-Y.L.)
| | - Chih-Yuan Lin
- Division of Cardiovascular Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan;
- Department of Biochemistry, National Defense Medical Center, Taipei 114, Taiwan; (J.-Y.C.); (P.-L.Y.); (P.-Z.L.); (S.-M.H.)
- Correspondence: (C.-S.T.); (C.-Y.L.); Tel.: +886-2-8792-7212 (C.-Y.L.); Fax: +886-2-8792-7376 (C.-Y.L.)
| |
Collapse
|
19
|
Liao FX, Huang F, Ma WG, Qin KP, Xu PF, Wu YF, Wang H, Chang J, Yin ZS. The New Role of Sirtuin1 in Human Osteoarthritis Chondrocytes by Regulating Autophagy. Cartilage 2021; 13:1237S-1248S. [PMID: 31072129 PMCID: PMC8804807 DOI: 10.1177/1947603519847736] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE The aim of this study is to investigate the role of Sirtuin1 (Sirt1) in the regulation of autophagy for human osteoarthritis (OA) chondrocytes. DESIGN All cartilage samples were collected from human donors, including young group, aged group, and OA group. Primary chondrocytes were isolated and cultured with Sirt1 activator or inhibitor. Sirt1 expression in cartilage tissue and chondrocytes was evaluated, and the deacetylation activity of Sirt1 was determined. The alteration of autophagy activity after upregulating or downregulating Sirt1 was detected. Chondrocytes were treated with autophagy activator and inhibitor, and then the protein level of Sirt1 was examined. The interactions between Sirt1 and autophagy-related proteins Atg7, microtubule associated protein 1 light chain 3 (LC3), and Beclin-1 were determined by using immunoprecipitation. RESULTS The assay of articular cartilage revealed that the expression of Sirt1 might be age-related: highly expressed in of younger people, and respectively decreased in the elderly people and OA patients. In vitro study was also validated this result. Further study confirmed that higher levels of Sirt1 significantly increased autophagy in aged chondrocytes, while the lower expression of Sirt1 reduced autophagy in young chondrocytes. Of note, the high levels of Sirt1 reduced autophagy in OA chondrocytes. When the chondrocytes were treated with autophagy activator or inhibitor, we found the expression of Sirt1 was not affected. In addition, we found that Sirt1 could interact with Atg7. CONCLUSION These results suggest that Sirt1 in human chondrocytes regulates autophagy by interacting with autophagy related Atg7, and Sirt1 may become a more important target in OA treatment.
Collapse
Affiliation(s)
- Fa-Xue Liao
- Department of Orthopaedics, The First
Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, People’s
Republic of China,Department of Orthopaedics, The Fourth
Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of
China
| | - Fei Huang
- Department of Orthopaedics, The Fourth
Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of
China
| | - Wen-Guang Ma
- Department of Orthopaedics, The Fourth
Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of
China
| | - Kun-Peng Qin
- Department of Orthopaedics, The Fourth
Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of
China
| | - Peng-Fei Xu
- Department of Orthopaedics, The Fourth
Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of
China
| | - Yun-Feng Wu
- Department of Orthopaedics, The Fourth
Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of
China
| | - Hao Wang
- Department of Orthopaedics, The Fourth
Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of
China
| | - Jun Chang
- Department of Orthopaedics, The Fourth
Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of
China
| | - Zong-Sheng Yin
- Department of Orthopaedics, The Fourth
Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of
China,Zong-Sheng Yin, Department of Orthopaedics,
The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road,
Hefei, Anhui Province 230022, China.
| |
Collapse
|
20
|
Maiese K. Neurodegeneration, memory loss, and dementia: the impact of biological clocks and circadian rhythm. FRONT BIOSCI-LANDMRK 2021; 26:614-627. [PMID: 34590471 PMCID: PMC8756734 DOI: 10.52586/4971] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/26/2021] [Accepted: 08/10/2021] [Indexed: 11/23/2022]
Abstract
Introduction: Dementia and cognitive loss impact a significant proportion of the global population and present almost insurmountable challenges for treatment since they stem from multifactorial etiologies. Innovative avenues for treatment are highly warranted. Methods and results: Novel work with biological clock genes that oversee circadian rhythm may meet this critical need by focusing upon the pathways of the mechanistic target of rapamycin (mTOR), the silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1), mammalian forkhead transcription factors (FoxOs), the growth factor erythropoietin (EPO), and the wingless Wnt pathway. These pathways are complex in nature, intimately associated with autophagy that can maintain circadian rhythm, and have an intricate relationship that can lead to beneficial outcomes that may offer neuroprotection, metabolic homeostasis, and prevention of cognitive loss. However, biological clocks and alterations in circadian rhythm also have the potential to lead to devastating effects involving tumorigenesis in conjunction with pathways involving Wnt that oversee angiogenesis and stem cell proliferation. Conclusions: Current work with biological clocks and circadian rhythm pathways provide exciting possibilities for the treating dementia and cognitive loss, but also provide powerful arguments to further comprehend the intimate and complex relationship among these pathways to fully potentiate desired clinical outcomes.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, New York, NY 10022, USA
| |
Collapse
|
21
|
Dimeric Histidine as a Novel Free Radical Scavenger Alleviates Non-Alcoholic Liver Injury. Antioxidants (Basel) 2021; 10:antiox10101529. [PMID: 34679664 PMCID: PMC8532998 DOI: 10.3390/antiox10101529] [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] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 09/22/2021] [Accepted: 09/24/2021] [Indexed: 01/20/2023] Open
Abstract
Non-alcoholic liver injury (NLI) is a common disease worldwide. Since free radical damage in the liver is a crucial initiator leading to diseases, scavenging excess free radicals has become an essential therapeutic strategy. To enhance the antioxidant capacity of histidine, we synthesized a protonated dimeric histidine, H-bihistidine, and investigated its anti-free radical potential in several free-radical-induced NLI. Results showed that H-bihistidine could strongly scavenge free radicals caused by H2O2, fatty acid, and CCl4, respectively, and recover cell viability in cultured hepatocytes. In the animal model of nonalcoholic fatty liver injury caused by high-fat diet, H-bihistidine reduced the contents of transaminases and lipids in serum, eliminated the liver’s fat accumulation, and decreased the oxidative damage. Moreover, H-bihistidine could rescue CCl4-induced liver injury and recover energy supply through scavenging free radicals. Moreover, liver fibrosis prepared by high-fat diet and CCl4 administration was significantly alleviated after H-bihistidine treatment. This study suggests a novel nonenzymatic free radical scavenger against NLI and, potentially, other free-radical-induced diseases.
Collapse
|
22
|
Maiese K. Cognitive Impairment and Dementia: Gaining Insight through Circadian Clock Gene Pathways. Biomolecules 2021; 11:1002. [PMID: 34356626 PMCID: PMC8301848 DOI: 10.3390/biom11071002] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/05/2021] [Accepted: 07/07/2021] [Indexed: 01/18/2023] Open
Abstract
Neurodegenerative disorders affect fifteen percent of the world's population and pose a significant financial burden to all nations. Cognitive impairment is the seventh leading cause of death throughout the globe. Given the enormous challenges to treat cognitive disorders, such as Alzheimer's disease, and the inability to markedly limit disease progression, circadian clock gene pathways offer an exciting strategy to address cognitive loss. Alterations in circadian clock genes can result in age-related motor deficits, affect treatment regimens with neurodegenerative disorders, and lead to the onset and progression of dementia. Interestingly, circadian pathways hold an intricate relationship with autophagy, the mechanistic target of rapamycin (mTOR), the silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1), mammalian forkhead transcription factors (FoxOs), and the trophic factor erythropoietin. Autophagy induction is necessary to maintain circadian rhythm homeostasis and limit cortical neurodegenerative disease, but requires a fine balance in biological activity to foster proper circadian clock gene regulation that is intimately dependent upon mTOR, SIRT1, FoxOs, and growth factor expression. Circadian rhythm mechanisms offer innovative prospects for the development of new avenues to comprehend the underlying mechanisms of cognitive loss and forge ahead with new therapeutics for dementia that can offer effective clinical treatments.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, New York, NY 10022, USA
| |
Collapse
|
23
|
Flessa CM, Kyrou I, Nasiri-Ansari N, Kaltsas G, Papavassiliou AG, Kassi E, Randeva HS. Endoplasmic Reticulum Stress and Autophagy in the Pathogenesis of Non-alcoholic Fatty Liver Disease (NAFLD): Current Evidence and Perspectives. Curr Obes Rep 2021; 10:134-161. [PMID: 33751456 DOI: 10.1007/s13679-021-00431-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/23/2021] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Non-alcoholic fatty liver disease (NAFLD) is one of the most common causes of chronic liver disease with rising prevalence worldwide. Herein, we provide a comprehensive overview of the current knowledge supporting the role of ER stress and autophagy processes in NAFLD pathogenesis and progression. We also highlight the interrelation between these two pathways and the impact of ER stress and autophagy modulators on NAFLD treatment. RECENT FINDINGS The pathophysiological mechanisms involved in NAFLD progression are currently under investigation. The endoplasmic reticulum (ER) stress and the concomitant unfolded protein response (UPR) seem to contribute to its pathogenesis mainly due to high ER content in the liver which exerts significant metabolic functions and can be dysregulated. Furthermore, disruption of autophagy processes has also been identified in NAFLD. The crucial role of these two pathways in NAFLD is underlined by the fact that they have recently emerged as promising targets of therapeutic interventions. There is a greater need for finding the natural/chemical compounds and drugs which can modulate the ER stress pathway and autophagy for the treatment of NAFLD. Clarifying the inter-relation between these two pathways and their interaction with inflammatory and apoptotic mechanisms will allow the development of additional therapeutic options which can better target and reprogram the underlying pathophysiological pathways, aiming to attenuate NAFLD progression.
Collapse
Affiliation(s)
- Christina-Maria Flessa
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, UK
| | - Ioannis Kyrou
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, UK
- Aston Medical Research Institute, Aston Medical School, College of Health and Life Sciences, Aston University, B4 7ET, Birmingham, UK
- Division of Translational and Experimental Medicine, Metabolic and Vascular Health, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
| | - Narjes Nasiri-Ansari
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | - Gregory Kaltsas
- Endocrine Unit, 1st Department of Propaedeutic and Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | - Athanasios G Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | - Eva Kassi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece.
- Endocrine Unit, 1st Department of Propaedeutic and Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, 11527, Athens, Greece.
| | - Harpal S Randeva
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, UK.
- Division of Translational and Experimental Medicine, Metabolic and Vascular Health, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK.
| |
Collapse
|
24
|
Zhao M, Chen S, Ji X, Shen X, You J, Liang X, Yin H, Zhao L. Current innovations in nutraceuticals and functional foods for intervention of non-alcoholic fatty liver disease. Pharmacol Res 2021; 166:105517. [PMID: 33636349 DOI: 10.1016/j.phrs.2021.105517] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/27/2021] [Accepted: 02/21/2021] [Indexed: 02/07/2023]
Abstract
As innovations in global agricultural production and food trading systems lead to major dietary shifts, high morbidity rates from non-alcoholic fatty liver disease (NAFLD), accompanied by elevated risk of lipid metabolism-related complications, has emerged as a growing problem worldwide. Treatment and prevention of NAFLD and chronic liver disease depends on the availability of safe, effective, and diverse therapeutic agents, the development of which is urgently needed. Supported by a growing body of evidence, considerable attention is now focused on interventional approaches that combines nutraceuticals and functional foods. In this review, we summarize the pathological progression of NAFLD and discuss the beneficial effects of nutraceuticals and the active ingredients in functional foods. We also describe the underlying mechanisms of these compounds in the intervention of NAFLD, including their effects on regulation of lipid homeostasis, activation of signaling pathways, and their role in gut microbial community dynamics and the gut-liver axis. In order to identify novel targets for treatment of lipid metabolism-related diseases, this work broadly explores the molecular mechanism linking nutraceuticals and functional foods, host physiology, and gut microbiota. Additionally, the limitations in existing knowledge and promising research areas for development of active interventions and treatments against NAFLD are discussed.
Collapse
Affiliation(s)
- Mengyao Zhao
- School of Biotechnology, State Key Laboratory of Bioreactor Engineering, R&D Center of Separation and Extraction Technology in Fermentation Industry, East China University of Science and Technology, Shanghai 200237, China; Shanghai Collaborative Innovation Center for Biomanufacturing Technology (SCICBT), Shanghai 200237, China
| | - Shumin Chen
- School of Biotechnology, State Key Laboratory of Bioreactor Engineering, R&D Center of Separation and Extraction Technology in Fermentation Industry, East China University of Science and Technology, Shanghai 200237, China
| | - Xiaoguo Ji
- School of Biotechnology, State Key Laboratory of Bioreactor Engineering, R&D Center of Separation and Extraction Technology in Fermentation Industry, East China University of Science and Technology, Shanghai 200237, China
| | - Xin Shen
- School of Biotechnology, State Key Laboratory of Bioreactor Engineering, R&D Center of Separation and Extraction Technology in Fermentation Industry, East China University of Science and Technology, Shanghai 200237, China
| | - Jiangshan You
- School of Biotechnology, State Key Laboratory of Bioreactor Engineering, R&D Center of Separation and Extraction Technology in Fermentation Industry, East China University of Science and Technology, Shanghai 200237, China
| | - Xinyi Liang
- School of Biotechnology, State Key Laboratory of Bioreactor Engineering, R&D Center of Separation and Extraction Technology in Fermentation Industry, East China University of Science and Technology, Shanghai 200237, China
| | - Hao Yin
- Organ Transplant Center, Shanghai Changzheng Hospital, Shanghai 200003, China.
| | - Liming Zhao
- School of Biotechnology, State Key Laboratory of Bioreactor Engineering, R&D Center of Separation and Extraction Technology in Fermentation Industry, East China University of Science and Technology, Shanghai 200237, China; School of Life Science, Shandong University of Technology, Zibo, Shandong 255000, China; Shanghai Collaborative Innovation Center for Biomanufacturing Technology (SCICBT), Shanghai 200237, China.
| |
Collapse
|
25
|
Role of NAD + in regulating cellular and metabolic signaling pathways. Mol Metab 2021; 49:101195. [PMID: 33609766 PMCID: PMC7973386 DOI: 10.1016/j.molmet.2021.101195] [Citation(s) in RCA: 189] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/02/2021] [Accepted: 02/15/2021] [Indexed: 12/15/2022] Open
Abstract
Background Nicotinamide adenine dinucleotide (NAD+), a critical coenzyme present in every living cell, is involved in a myriad of metabolic processes associated with cellular bioenergetics. For this reason, NAD+ is often studied in the context of aging, cancer, and neurodegenerative and metabolic disorders. Scope of review Cellular NAD+ depletion is associated with compromised adaptive cellular stress responses, impaired neuronal plasticity, impaired DNA repair, and cellular senescence. Increasing evidence has shown the efficacy of boosting NAD+ levels using NAD+ precursors in various diseases. This review provides a comprehensive understanding into the role of NAD+ in aging and other pathologies and discusses potential therapeutic targets. Major conclusions An alteration in the NAD+/NADH ratio or the NAD+ pool size can lead to derailment of the biological system and contribute to various neurodegenerative disorders, aging, and tumorigenesis. Due to the varied distribution of NAD+/NADH in different locations within cells, the direct role of impaired NAD+-dependent processes in humans remains unestablished. In this regard, longitudinal studies are needed to quantify NAD+ and its related metabolites. Future research should focus on measuring the fluxes through pathways associated with NAD+ synthesis and degradation. NAD+ regulates energy metabolism, DNA damage repair, gene expression, and stress response. NAD+ deterioration contributes to the progression of multiple metabolic disorders, cancers, and neurodegenerative diseases. Nicotinamide mononucleotide and nicotinamide riboside raise NAD+ levels in different tissues in preclinical models. Imaging studies on genetic models can illustrate the pathways of NAD+metabolism and their downstream functional effects. Human clinical trials to determine benefits of restoration of NAD+ by using NAD precursors are in progress.
Collapse
|
26
|
Xu T, Song Q, Zhou L, Yang W, Wu X, Qian Q, Chai H, Han Q, Pan H, Dou X, Li S. Ferulic acid alleviates lipotoxicity-induced hepatocellular death through the SIRT1-regulated autophagy pathway and independently of AMPK and Akt in AML-12 hepatocytes. Nutr Metab (Lond) 2021; 18:13. [PMID: 33468182 PMCID: PMC7814733 DOI: 10.1186/s12986-021-00540-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 01/02/2021] [Indexed: 02/07/2023] Open
Abstract
Background Lipotoxicity-induced cell death plays a detrimental role in the pathogenesis of metabolic diseases. Ferulic acid, widespread in plant-based food, is a radical scavenger with multiple bioactivities. However, the benefits of ferulic acid against hepatic lipotoxicity are largely unclear. Here, we investigated the protective effect of ferulic acid against palmitate-induced lipotoxicity and clarified its potential mechanisms in AML-12 hepatocytes. Methods AML-12 mouse hepatocytes were exposed to palmitate to mimic lipotoxicity. Different doses (25, 50, and 100 μM) of ferulic acid were added 2 h before palmitate treatment. Cell viability was detected by measuring lactate dehydrogenase release, nuclear staining, and the expression of cleaved-caspase-3. Intracellular reactive oxygen species content and mitochondrial membrane potential were analysed by fluorescent probes. The potential mechanisms were explored by molecular biological methods, including Western blotting and quantitative real-time PCR, and were further verified by siRNA interference. Results Our data showed that ferulic acid significantly inhibited palmitate-induced cell death, rescued mitochondrial membrane potential, reduced reactive oxygen species accumulation, and decreased inflammatory factor activation, including IL-6 and IL-1beta. Ferulic acid significantly stimulated autophagy in hepatocytes, whereas autophagy suppression blocked the protective effect of ferulic acid against lipotoxicity. Ferulic acid-activated autophagy, which was triggered by SIRT1 upregulation, was mechanistically involved in its anti-lipotoxicity effects. SIRT1 silencing blocked most beneficial changes induced by ferulic acid. Conclusions We demonstrated that the phytochemical ferulic acid, which is found in plant-based food, protected against hepatic lipotoxicity, through the SIRT1/autophagy pathway. Increased intake of ferulic acid-enriched food is a potential strategy to prevent and/or improve metabolic diseases with lipotoxicity as a typical pathological feature.
Collapse
Affiliation(s)
- Tiantian Xu
- College of Basic Medicine and Public Health, Zhejiang Chinese Medical University, Hangzhou, 310053, China.,College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Qing Song
- College of Basic Medicine and Public Health, Zhejiang Chinese Medical University, Hangzhou, 310053, China.,College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China.,Molecular Medicine Institute, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Li Zhou
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China.,The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Wenwen Yang
- College of Basic Medicine and Public Health, Zhejiang Chinese Medical University, Hangzhou, 310053, China.,College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xiangyao Wu
- College of Basic Medicine and Public Health, Zhejiang Chinese Medical University, Hangzhou, 310053, China.,College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Qianyu Qian
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China.,Molecular Medicine Institute, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Hui Chai
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China.,Molecular Medicine Institute, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Qiang Han
- College of Basic Medicine and Public Health, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Hongzhi Pan
- Collaborative Research Center, Shanghai University of Medicine and Health Sciences, Shanghai, 201399, China
| | - Xiaobing Dou
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China. .,Molecular Medicine Institute, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Songtao Li
- College of Basic Medicine and Public Health, Zhejiang Chinese Medical University, Hangzhou, 310053, China. .,Molecular Medicine Institute, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
27
|
Chen H, Chen F, Zhang M, Chen Y, Cui L, Liang C. A Review of APOE Genotype-Dependent Autophagic Flux Regulation in Alzheimer's Disease. J Alzheimers Dis 2021; 84:535-555. [PMID: 34569952 DOI: 10.3233/jad-210602] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Autophagy is a basic physiological process maintaining cell renewal, the degradation of dysfunctional organelles, and the clearance of abnormal proteins and has recently been identified as a main mechanism underlying the onset and progression of Alzheimer's disease (AD). The APOE ɛ4 genotype is the strongest genetic determinant of AD pathogenesis and initiates autophagic flux at different times. This review synthesizes the current knowledge about the potential pathogenic effects of ApoE4 on autophagy and describes its associations with the biological hallmarks of autophagy and AD from a novel perspective. Via a remarkable variety of widely accepted signaling pathway markers, such as mTOR, TFEB, SIRT1, LC3, p62, LAMP1, LAMP2, CTSD, Rabs, and V-ATPase, ApoE isoforms differentially modulate autophagy initiation; membrane expansion, recruitment, and enclosure; autophagosome and lysosome fusion; and lysosomal degradation. Although the precise pathogenic mechanism varies for different genes and proteins, the dysregulation of autophagic flux is a key mechanism on which multiple pathogenic processes converge.
Collapse
Affiliation(s)
- Huiyi Chen
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Yuebei People's Hospital, Affiliated Hospital of Shantou University Medical College, Shaoguan, China
| | - Feng Chen
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Miaoping Zhang
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yanting Chen
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Lili Cui
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Chunmei Liang
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
28
|
Liu N, Zeng L, Zhang YM, Pan W, Lai H. Astaxanthin alleviates pathological brain aging through the upregulation of hippocampal synaptic proteins. Neural Regen Res 2021; 16:1062-1067. [PMID: 33269751 PMCID: PMC8224122 DOI: 10.4103/1673-5374.300460] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Oxidative stress is currently considered to be the main cause of brain aging. Astaxanthin can improve oxidative stress under multiple pathological conditions. It is therefore hypothesized that astaxanthin might have therapeutic effects on brain aging. To validate this hypothesis and investigate the underlying mechanisms, a mouse model of brain aging was established by injecting amyloid beta (Aβ)25–35 (5 μM, 3 μL/injection, six injections given every other day) into the right lateral ventricle. After 3 days of Aβ25–35 injections, the mouse models were intragastrically administered astaxanthin (0.1 mL/d, 10 mg/kg) for 30 successive days. Astaxanthin greatly reduced the latency to find the platform in the Morris water maze, increased the number of crossings of the target platform, and increased the expression of brain-derived neurotrophic factor, synaptophysin, sirtuin 1, and peroxisome proliferator-activated receptor-γ coactivator 1α. Intraperitoneal injection of the sirtuin 1 inhibitor nicotinamide (500 μM/d) for 7 successive days after astaxanthin intervention inhibited these phenomena. These findings suggest that astaxanthin can regulate the expression of synaptic proteins in mouse hippocampus through the sirtuin 1/peroxisome proliferator-activated receptor-γ coactivator 1α signaling pathway, which leads to improvements in the learning, cognitive, and memory abilities of mice. The study was approved by the Animal Ethics Committee, China Medical University, China (approval No. CMU2019294) on January 15, 2019.
Collapse
Affiliation(s)
- Ning Liu
- 1Department of Human Anatomy, College of Basic Medicine, China Medical University, Shenyang; Department of Radiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province, China
| | - Liang Zeng
- Department of Human Anatomy, College of Basic Medicine, Shenyang Medical College, Shenyang, Liaoning Province, China
| | - Yi-Ming Zhang
- Department of Human Anatomy, College of Basic Medicine, China Medical University, Shenyang, Liaoning Province, China
| | - Wang Pan
- Department of Neurobiology of Jinzhou Medical University, Jinzhou, Liaoning Province, China
| | - Hong Lai
- Department of Human Anatomy, College of Basic Medicine, China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
29
|
Maiese K. Nicotinamide as a Foundation for Treating Neurodegenerative Disease and Metabolic Disorders. Curr Neurovasc Res 2021; 18:134-149. [PMID: 33397266 PMCID: PMC8254823 DOI: 10.2174/1567202617999210104220334] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 02/06/2023]
Abstract
Neurodegenerative disorders impact more than one billion individuals worldwide and are intimately tied to metabolic disease that can affect another nine hundred individuals throughout the globe. Nicotinamide is a critical agent that may offer fruitful prospects for neurodegenerative diseases and metabolic disorders, such as diabetes mellitus. Nicotinamide protects against multiple toxic environments that include reactive oxygen species exposure, anoxia, excitotoxicity, ethanolinduced neuronal injury, amyloid (Aß) toxicity, age-related vascular disease, mitochondrial dysfunction, insulin resistance, excess lactate production, and loss of glucose homeostasis with pancreatic β-cell dysfunction. However, nicotinamide offers cellular protection in a specific concentration range, with dosing outside of this range leading to detrimental effects. The underlying biological pathways of nicotinamide that involve the silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1), the mechanistic target of rapamycin (mTOR), AMP activated protein kinase (AMPK), and mammalian forkhead transcription factors (FoxOs) may offer insight for the clinical translation of nicotinamide into a safe and efficacious therapy through the modulation of oxidative stress, apoptosis, and autophagy. Nicotinamide is a highly promising target for the development of innovative strategies for neurodegenerative disorders and metabolic disease, but the benefits of this foundation depend greatly on gaining a further understanding of nicotinamide's complex biology.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, New York, New York 10022
| |
Collapse
|
30
|
Chen Y, Griffiths A, Wang J, Zhang T, Song Q, Song Z. Inositol-requiring enzyme 1α links palmitate-induced mTOR activation and lipotoxicity in hepatocytes. Am J Physiol Cell Physiol 2020; 319:C1130-C1140. [PMID: 33052067 PMCID: PMC7792676 DOI: 10.1152/ajpcell.00165.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 10/05/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023]
Abstract
Hepatic lipotoxicity, hepatocyte dysfunction/cell death induced by saturated fatty acids (SFA), plays a central role in the pathogenesis of nonalcoholic fatty liver disease (NAFLD); however, the underlying mechanisms remain unclear. Palmitate is the most abundant SFA in the circulation. In this study, via a small-scale screening of chemical inhibitors using AML12 hepatocytes, we identified mechanistic target of rapamycin (mTOR) complex 1 (mTORC1) to be a culprit in palmitate-induced cell death in hepatocytes in that mTOR inhibition is protective against palmitate-induced cell death. The protective effect of mTORC1 inhibition is independent of autophagy induction, as autophagy inhibition failed to ablate the mTORC1 inhibitor-conferred protection. We have previously reported that the endonuclease activity of inositol-requiring enzyme 1α (IRE1α), one of three canonical signaling pathways of endoplasmic reticulum (ER) stress, was implicated in palmitate-induced cell death in hepatocytes. The continuous mechanistic investigation in this study uncovered that IRE1α is a downstream target of mTORC1 activation upon palmitate exposure and the inhibition of either its endonuclease activity or kinase activity protects against the lipotoxic effect of palmitate. Our research further revealed that protein palmitoylation is potentially involved in palmitate-induced mTORC1 activation and lipotoxicity in hepatocytes. 2-Bromopalmitate, a protein palmitoylation inhibitor, ameliorated palmitate-triggered mTORC1 activation, concomitant with the protection of lipotoxicity in hepatocytes. Collectively, our data have identified that mTORC1 and ER stress are coordinately implicated in hepatocyte cell death in response to palmitate exposure and suggest that this pathway may potentially serve as a therapeutic target for the treatment of NAFLD as well as other metabolic disorders involving lipotoxicity.
Collapse
Affiliation(s)
- Yingli Chen
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois
- College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, Heilongjiang, People's Republic of China
| | - Alexandra Griffiths
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois
| | - Jun Wang
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois
- Department of Gastroenterology, Tongji Medical College and Central Hospital of Wuhan, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Tingting Zhang
- Department of Urology, Daqing Oilfield General Hospital, Daqing, Heilongjiang, People's Republic of China
| | - Qing Song
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois
| | - Zhenyuan Song
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
31
|
Li S, Qian Q, Ying N, Lai J, Feng L, Zheng S, Jiang F, Song Q, Chai H, Dou X. Activation of the AMPK-SIRT1 pathway contributes to protective effects of Salvianolic acid A against lipotoxicity in hepatocytes and NAFLD in mice. Front Pharmacol 2020; 11:560905. [PMID: 33328983 PMCID: PMC7734334 DOI: 10.3389/fphar.2020.560905] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 09/30/2020] [Indexed: 12/15/2022] Open
Abstract
Background: Salvianolic acid A (Sal A), a natural polyphenol compound extracted from Radix Salvia miltiorrhiza (known as Danshen in China), possesses a variety of potential pharmacological activities. The aim of this study is to determine mechanisms of hepatoprotective effects of Sal A against lipotoxicity both in cultured hepatocytes and in a mouse model of fatty liver disease. Methods: High-fat and high-carbohydrate diet (HFCD)-fed C57BL/6J mice were employed to establish hepatic lipotoxicity in a mouse model. Two doses of Sal A were administered every other day via intraperitoneal injection (20 and 40 mg/kg BW, respectively). After a 10-week intervention, liver injury was detected by immunohistochemical and biochemical analyses. For in vitro studies, we used HepG2, a human hepatoma cell line, and exposed them to palmitic acid to induce lipotoxicity. The protective effects of Sal A on palmitic acid-induced lipotoxicity were examined in Sal A-pretreated HepG2 cells. Results: Sal A treatments attenuated body weight gain, liver injury, and hepatic steatosis in mice exposed to HFCD. Sal A pretreatments ameliorated palmitic acid-induced cell death but did not reverse effects of HFCD- or palmitate-induced activations of JNK, ERK1/2, and PKA. Induction of p38 phosphorylation was significantly reversed by Sal A in HFCD-fed mice but not in palmitate-treated HepG2 cells. However, Sal A rescued hepatic AMP-activated protein kinase (AMPK) suppression and sirtuin 1 (SIRT1) downregulation by both HFCD feeding in mice and exposure to palmitate in HepG2 cells. Sal A dose-dependently up-regulated p-AMPK and SIRT1 protein levels. Importantly, siRNA silencing of either AMPK or SIRT1 gene expression abolished the protective effects of Sal A on lipotoxicity. Moreover, while AMPK silencing blocked Sal A-induced SIRT1, silencing of SIRT1 had no effect on Sal A-triggered AMPK activation, suggesting SIRT1 upregulation by Sal A is mediated by AMPK activation. Conclusion: Our data uncover a novel mechanism for hepatoprotective effects of Sal A against lipotoxicity both in livers from HFCD-fed mice and palmitic acid-treated hepatocytes.
Collapse
Affiliation(s)
- Songtao Li
- College of Basic Medicine and Public Health, Zhejiang Chinese Medical University, Hangzhou, China
- Molecular Medicine Institute, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qianyu Qian
- Molecular Medicine Institute, Zhejiang Chinese Medical University, Hangzhou, China
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Na Ying
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jianfei Lai
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Luyan Feng
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Sitong Zheng
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fusheng Jiang
- Molecular Medicine Institute, Zhejiang Chinese Medical University, Hangzhou, China
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qing Song
- Molecular Medicine Institute, Zhejiang Chinese Medical University, Hangzhou, China
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Hui Chai
- Molecular Medicine Institute, Zhejiang Chinese Medical University, Hangzhou, China
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaobing Dou
- Molecular Medicine Institute, Zhejiang Chinese Medical University, Hangzhou, China
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
32
|
Cha SH, Hwang Y, Heo SJ, Jun HS. Diphlorethohydroxycarmalol Attenuates Palmitate-Induced Hepatic Lipogenesis and Inflammation. Mar Drugs 2020; 18:E475. [PMID: 32962167 PMCID: PMC7551772 DOI: 10.3390/md18090475] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/11/2020] [Accepted: 09/15/2020] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a common cause of chronic liver disease, encompassing a range of conditions caused by lipid deposition within liver cells, and is also associated with obesity and metabolic diseases. Here, we investigated the protective effects of diphlorethohydroxycarmalol (DPHC), which is a polyphenol isolated from an edible seaweed, Ishige okamurae, on palmitate-induced lipotoxicity in the liver. DPHC treatment repressed palmitate-induced cytotoxicity, triglyceride content, and lipid accumulation. DPHC prevented palmitate-induced mRNA and protein expression of SREBP (sterol regulatory element-binding protein) 1, C/EBP (CCAAT-enhancer-binding protein) α, ChREBP (carbohydrate-responsive element-binding protein), and FAS (fatty acid synthase). In addition, palmitate treatment reduced the expression levels of phosphorylated AMP-activated protein kinase (AMPK) and sirtuin (SIRT)1 proteins, and DPHC treatment rescued this reduction. Moreover, DPHC protected palmitate-induced liver toxicity and lipogenesis, as well as inflammation, and enhanced AMPK and SIRT1 signaling in zebrafish. These results suggest that DPHC possesses protective effects against palmitate-induced toxicity in the liver by preventing lipogenesis and inflammation. DPHC could be used as a potential therapeutic or preventive agent for fatty liver diseases.
Collapse
Affiliation(s)
- Seon-Heui Cha
- Department of Marine Bio and Medical Sciences, Hanseo University, Chungcheongnam-do 31962, Korea
| | - Yongha Hwang
- Gachon Medical and Convergence Institute, Gachon Gil Medical Center, Incheon 21999, Korea;
| | - Soo-Jin Heo
- Jeju Marine Research Center, Korea Institute of Ocean Science and Technology (KIOST), Jeju 63349, Korea;
- Department of Biology, University of Science and Technology (UST), Daejeon 34113, Korea
| | - Hee-Sook Jun
- Gachon Medical and Convergence Institute, Gachon Gil Medical Center, Incheon 21999, Korea;
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Korea
- College of Pharmacy, Gachon University, Incheon 21999, Korea
| |
Collapse
|
33
|
Kitaoka Y, Sase K, Tsukahara C, Fujita N, Arizono I, Takagi H. Axonal Protection by Nicotinamide Riboside via SIRT1-Autophagy Pathway in TNF-Induced Optic Nerve Degeneration. Mol Neurobiol 2020; 57:4952-4960. [PMID: 32820458 PMCID: PMC7541376 DOI: 10.1007/s12035-020-02063-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 08/07/2020] [Indexed: 02/07/2023]
Abstract
Nicotinamide adenine dinucleotide (NAD+) synthesis pathway has been involved in many biological functions. Nicotinamide riboside (NR) is widely used as an NAD+ precursor and known to increase NAD+ level in several tissues. The present study aimed to examine the effect of NR on tumor necrosis factor (TNF)-induced optic nerve degeneration and to investigate whether it alters SIRT1 expression and autophagic status in optic nerve. We also examined the localization of nicotinamide riboside kinase 1 (NRK1), which is a downstream enzyme for NR biosynthesis pathway in retina and optic nerve. Intravitreal injection of TNF or TNF plus NR was performed on rats. The p62 and LC3-II protein levels were examined to evaluate autophagic flux in optic nerve. Immunohistochemical analysis was performed to localize NRK1 expression. Morphometric analysis showed substantial axonal protection by NR against TNF-induced axon loss. TNF-induced increment of p62 protein level was significantly inhibited by NR administration. NR administration alone significantly increased the LC3-II levels and reduced p62 levels compared with the basal levels, and upregulated SIRT1 levels in optic nerve. Immunohistochemical analysis showed that NRK1 exists in retinal ganglion cells (RGCs) and nerve fibers in retina and optic nerve. NR administration apparently upregulated NRK1 levels in the TNF-treated eyes as well as the control eyes. Pre-injection of an SIRT1 inhibitor resulted in a significant increase of p62 levels in the NR plus TNF treatment group, implicating that SIRT1 regulates autophagy status. In conclusion, NRK1 exists in RGCs and optic nerve axons. NR exerted protection against axon loss induced by TNF with possible involvement of upregulated NRK1 and SIRT1-autophagy pathway.
Collapse
Affiliation(s)
- Yasushi Kitaoka
- Department of Molecular Neuroscience, St. Marianna University Graduate School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kaswasaki, Kanagawa, 216-8511, Japan.
| | - Kana Sase
- Department of Ophthalmology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Chihiro Tsukahara
- Department of Molecular Neuroscience, St. Marianna University Graduate School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kaswasaki, Kanagawa, 216-8511, Japan.,Department of Ophthalmology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Naoki Fujita
- Department of Molecular Neuroscience, St. Marianna University Graduate School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kaswasaki, Kanagawa, 216-8511, Japan.,Department of Ophthalmology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Ibuki Arizono
- Department of Molecular Neuroscience, St. Marianna University Graduate School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kaswasaki, Kanagawa, 216-8511, Japan.,Department of Ophthalmology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Hitoshi Takagi
- Department of Ophthalmology, St. Marianna University School of Medicine, Kawasaki, Japan
| |
Collapse
|
34
|
Buqué A, Bloy N, Kroemer G, Galluzzi L. Possible mechanisms of cancer prevention by nicotinamide. Br J Pharmacol 2020; 178:2034-2040. [DOI: 10.1111/bph.15096] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 04/21/2020] [Accepted: 04/22/2020] [Indexed: 12/13/2022] Open
Affiliation(s)
- Aitziber Buqué
- Department of Radiation Oncology Weill Cornell Medical College New York NY USA
| | - Norma Bloy
- Department of Radiation Oncology Weill Cornell Medical College New York NY USA
| | - Guido Kroemer
- Equipe labellisée par la Ligue contre le cancer Université de Paris, Sorbonne Université, INSERM U1138, Centre de Recherche des Cordeliers Paris France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP‐HP Paris France
- Suzhou Institute for Systems Medicine Chinese Academy of Sciences Suzhou China
- Department of Women's and Children's Health Karolinska University Hospital Stockholm Sweden
| | - Lorenzo Galluzzi
- Department of Radiation Oncology Weill Cornell Medical College New York NY USA
- Sandra and Edward Meyer Cancer Center New York NY USA
- Caryl and Israel Englander Institute for Precision Medicine New York NY USA
- Department of Dermatology Yale School of Medicine New Haven CT USA
- Université de Paris Paris France
| |
Collapse
|
35
|
O’Dwyer C, Yaworski R, Katsumura S, Ghorbani P, Gobeil Odai K, Nunes JR, LeBlond ND, Sanjana S, Smith TT, Han S, Margison KD, Alain T, Morita M, Fullerton MD. Hepatic Choline Transport Is Inhibited During Fatty Acid-Induced Lipotoxicity and Obesity. Hepatol Commun 2020; 4:876-889. [PMID: 32490323 PMCID: PMC7262319 DOI: 10.1002/hep4.1516] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 02/25/2020] [Accepted: 03/11/2020] [Indexed: 01/11/2023] Open
Abstract
Choline is an essential nutrient and a critical component of the membrane phospholipid phosphatidylcholine (PC), the neurotransmitter acetylcholine, while also contributing to the methylation pathway. In the liver specifically, PC is the major membrane constituent and can be synthesized by the cytidine diphosphate-choline or the phosphatidylethanolamine N-methyltransferase pathway. With the continuing global rise in the rates of obesity and nonalcoholic fatty liver disease, we sought to explore how excess fatty acids on primary hepatocytes and diet-induced obesity affect choline uptake and metabolism. Our results demonstrate that hepatocytes chronically treated with palmitate, but not oleate or a mixture, had decreased choline uptake, which was associated with lower choline incorporation into PC and lower expression of choline transport proteins. Interestingly, a reduction in the rate of degradation spared PC levels in response to palmitate when compared with control. The effects of palmitate treatment were independent of endoplasmic reticulum stress, which counterintuitively augmented choline transport and transporter expression. In a model of obesity-induced hepatic steatosis, male mice fed a 60% high-fat diet for 10 weeks had significantly diminished hepatic choline uptake compared with lean mice fed a control diet. Although the transcript and protein expression of various choline metabolic enzymes fluctuated slightly, we observed reduced protein expression of choline transporter-like 1 (CTL1) in the liver of mice fed a high-fat diet. Polysome profile analyses revealed that in livers of obese mice, the CTL1 transcript, despite being more abundant, was translated to a lesser extent compared with lean controls. Finally, human liver cells demonstrated a similar response to palmitate treatment. Conclusion: Our results suggest that the altered fatty acid milieu seen in obesity-induced fatty liver disease progression may adversely affect choline metabolism, potentially through CTL1, but that compensatory mechanisms work to maintain phospholipid homeostasis.
Collapse
Affiliation(s)
- Conor O’Dwyer
- Department of Biochemistry, Microbiology and ImmunologyFaculty of MedicineUniversity of OttawaOttawaONCanada
- Centre for InfectionImmunity and Inflammation and Centre for Catalysis Research and InnovationUniversity of OttawaOttawaONCanada
| | - Rebecca Yaworski
- Department of Biochemistry, Microbiology and ImmunologyFaculty of MedicineUniversity of OttawaOttawaONCanada
- Centre for InfectionImmunity and Inflammation and Centre for Catalysis Research and InnovationUniversity of OttawaOttawaONCanada
| | - Sakie Katsumura
- Department of Molecular MedicineUniversity of Texas Health Science Center at San AntonioSan AntonioTX
| | - Peyman Ghorbani
- Department of Biochemistry, Microbiology and ImmunologyFaculty of MedicineUniversity of OttawaOttawaONCanada
- Centre for InfectionImmunity and Inflammation and Centre for Catalysis Research and InnovationUniversity of OttawaOttawaONCanada
| | - Kaelan Gobeil Odai
- Department of Biochemistry, Microbiology and ImmunologyFaculty of MedicineUniversity of OttawaOttawaONCanada
- Centre for InfectionImmunity and Inflammation and Centre for Catalysis Research and InnovationUniversity of OttawaOttawaONCanada
| | - Julia R.C. Nunes
- Department of Biochemistry, Microbiology and ImmunologyFaculty of MedicineUniversity of OttawaOttawaONCanada
- Centre for InfectionImmunity and Inflammation and Centre for Catalysis Research and InnovationUniversity of OttawaOttawaONCanada
| | - Nicholas D. LeBlond
- Department of Biochemistry, Microbiology and ImmunologyFaculty of MedicineUniversity of OttawaOttawaONCanada
- Centre for InfectionImmunity and Inflammation and Centre for Catalysis Research and InnovationUniversity of OttawaOttawaONCanada
| | - Sabrin Sanjana
- Department of Biochemistry, Microbiology and ImmunologyFaculty of MedicineUniversity of OttawaOttawaONCanada
- Centre for InfectionImmunity and Inflammation and Centre for Catalysis Research and InnovationUniversity of OttawaOttawaONCanada
| | - Tyler T.K. Smith
- Department of Biochemistry, Microbiology and ImmunologyFaculty of MedicineUniversity of OttawaOttawaONCanada
- Centre for InfectionImmunity and Inflammation and Centre for Catalysis Research and InnovationUniversity of OttawaOttawaONCanada
| | - Shauna Han
- Department of Biochemistry, Microbiology and ImmunologyFaculty of MedicineUniversity of OttawaOttawaONCanada
- Centre for InfectionImmunity and Inflammation and Centre for Catalysis Research and InnovationUniversity of OttawaOttawaONCanada
| | - Kaitlyn D. Margison
- Department of Biochemistry, Microbiology and ImmunologyFaculty of MedicineUniversity of OttawaOttawaONCanada
- Centre for InfectionImmunity and Inflammation and Centre for Catalysis Research and InnovationUniversity of OttawaOttawaONCanada
| | - Tommy Alain
- Department of Biochemistry, Microbiology and ImmunologyFaculty of MedicineUniversity of OttawaOttawaONCanada
- Centre for InfectionImmunity and Inflammation and Centre for Catalysis Research and InnovationUniversity of OttawaOttawaONCanada
- Children’s Hospital of Eastern Ontario Research InstituteOttawaONCanada
| | - Masahiro Morita
- Department of Molecular MedicineUniversity of Texas Health Science Center at San AntonioSan AntonioTX
- Barshop Institute for Longevity and Aging StudiesUniversity of Texas Health Science Center at San AntonioSan AntonioTX
- Institute of Resource Development and AnalysisKumamoto UniversityKumamotoJapan
| | - Morgan D. Fullerton
- Department of Biochemistry, Microbiology and ImmunologyFaculty of MedicineUniversity of OttawaOttawaONCanada
- Centre for InfectionImmunity and Inflammation and Centre for Catalysis Research and InnovationUniversity of OttawaOttawaONCanada
| |
Collapse
|
36
|
Abstract
Metabolic disorders, such as diabetes mellitus (DM), are increasingly becoming significant risk factors for the health of the global population and consume substantial portions of the gross domestic product of all nations. Although conventional therapies that include early diagnosis, nutritional modification of diet, and pharmacological treatments may limit disease progression, tight serum glucose control cannot prevent the onset of future disease complications. With these concerns, novel strategies for the treatment of metabolic disorders that involve the vitamin nicotinamide, the mechanistic target of rapamycin (mTOR), mTOR Complex 1 (mTORC1), mTOR Complex 2 (mTORC2), AMP activated protein kinase (AMPK), and the cellular pathways of autophagy and apoptosis offer exceptional promise to provide new avenues of treatment. Oversight of these pathways can promote cellular energy homeostasis, maintain mitochondrial function, improve glucose utilization, and preserve pancreatic beta-cell function. Yet, the interplay among mTOR, AMPK, and autophagy pathways can be complex and affect desired clinical outcomes, necessitating further investigations to provide efficacious treatment strategies for metabolic dysfunction and DM.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, New York, New York 10022,
| |
Collapse
|
37
|
Zhang E, Yin S, Zhao C, Fan L, Hu H. Involvement of activation of PLIN5-Sirt1 axis in protective effect of glycycoumarin on hepatic lipotoxicity. Biochem Biophys Res Commun 2020; 528:7-13. [PMID: 32448510 DOI: 10.1016/j.bbrc.2020.05.072] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 05/11/2020] [Indexed: 02/09/2023]
Abstract
Licorice is a popular medicinal plant, and it has been used to treat various diseases, including liver diseases. Glycycoumarin (GCM) is a major coumarin compound isolated from licorice with favorable bioavailability property. Our previous studies have shown that GCM is capable of inhibiting lipoapoptosis in both cell culture and methionine-choline-defcient (MCD) diet-induced mouse model of non-alcoholic steatohepatitis (NASH) through mechanisms involving suppression of endoplasmic reticulum (ER) stress. Perilipin 5 (PLIN5), a newly identified lipid drop protein in the perilipin family, is highly expressed in oxidative tissues including the liver and is suggested to play an important role in protecting against hepatic lipotoxicity. Give the hepatoprotective role of PLIN5, we hypothesized that induction of PLIN5 might contribute to the hepatoprotective effect of GCM via mitigating ER stress and inflammatory responses. Results showed that PLIN5 and its downstream target Sirt1 were induced by GCM both in vitro and in vivo. Inhibition of either PLIN5 or Sirt1 led to significantly attenuated protective effect of GCM on palmitic acid (PA)-induced lipoapoptosis and inflammatory responses, supporting involvement of PLIN5-Sirt1 axis in the protective effect of GCM on hepatic lipotoxicity. The findings of the present study provide novel insight into the understanding of mechanisms underlying the hepatoprotective effect of GCM.
Collapse
Affiliation(s)
- Enxiang Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, China
| | - Shutao Yin
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, China
| | - Chong Zhao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, China
| | - Lihong Fan
- College of Veterinary Medicine, China Agricultural University, China.
| | - Hongbo Hu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, China.
| |
Collapse
|
38
|
Wang J, Chen Y, Song Q, Griffiths A, Song Z. mTORC1-IRE1α pathway activation contributes to palmitate-elicited triglyceride secretion and cell death in hepatocytes. Exp Biol Med (Maywood) 2020; 245:1268-1279. [PMID: 32436749 DOI: 10.1177/1535370220928276] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
IMPACT STATEMENT Lipotoxicity induced by saturated fatty acids (SFA) plays a pivotal role in the pathogenesis of a variety of obesity-related metabolic disorders; however, the exact mechanism(s) underlying lipotoxicity development remains elusive. The liver plays a central role in regulating intrahepatic and circulatory lipid homeostasis. In the current study, we identified that mammalian target of rapamycin complex 1 (mTORC1) activation plays an important role in regulating the detrimental effects of SFA palmitate in hepatocytes, in specific cell death, and TG overproduction. Furthermore, our data confirmed that palmitate-induced mTORC1 activation is attributable to its stimulatory effect on IRE1α, one of three canonical pathways activated during ER stress. Importantly, IRE1α inhibition prevented palmitate-triggered cell death and TG overproduction, suggesting mTORC1-IRE1α pathway is mechanistically implicated in palmitate lipotoxicity. The data obtained in the current investigation support future study to explore the therapeutic potential of targeting the mTORC1-IRE1α pathway as a novel clinical strategy for the treatment of metabolic disorders involving lipotoxicity.
Collapse
Affiliation(s)
- Jun Wang
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612, USA.,Department of Gastroenterology, Tongji Medical College and The Central Hospital of Wuhan, Huazhong University of Science and Technology, Wuhan 430000, PR. China
| | - Yingli Chen
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612, USA.,College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, Heilongjiang 163319, PR. China
| | - Qing Song
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Alexandra Griffiths
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Zhenyuan Song
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
39
|
Hsieh CL, Hsieh SY, Huang HM, Lu SL, Omori H, Zheng PX, Ho YN, Cheng YL, Lin YS, Chiang-Ni C, Tsai PJ, Wang SY, Liu CC, Noda T, Wu JJ. Nicotinamide Increases Intracellular NAD + Content to Enhance Autophagy-Mediated Group A Streptococcal Clearance in Endothelial Cells. Front Microbiol 2020; 11:117. [PMID: 32117141 PMCID: PMC7026195 DOI: 10.3389/fmicb.2020.00117] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 01/20/2020] [Indexed: 12/16/2022] Open
Abstract
Group A streptococcus (GAS) is a versatile pathogen that causes a wide spectrum of diseases in humans. Invading host cells is a known strategy for GAS to avoid antibiotic killing and immune recognition. However, the underlying mechanisms of GAS resistance to intracellular killing need to be explored. Endothelial HMEC-1 cells were infected with GAS, methicillin-resistant Staphylococcus aureus (MRSA) and Salmonella Typhimurium under nicotinamide (NAM)-supplemented conditions. The intracellular NAD+ level and cell viability were respectively measured by NAD+ quantification kit and protease-based cytotoxicity assay. Moreover, the intracellular bacteria were analyzed by colony-forming assay, transmission electron microscopy, and confocal microscopy. We found that supplementation with exogenous nicotinamide during infection significantly inhibited the growth of intracellular GAS in endothelial cells. Moreover, the NAD+ content and NAD+/NADH ratio of GAS-infected endothelial cells were dramatically increased, whereas the cell cytotoxicity was decreased by exogenous nicotinamide treatment. After knockdown of the autophagy-related ATG9A, the intracellular bacterial load was increased in nicotinamide-treated endothelial cells. The results of Western blot and transmission electron microscopy also revealed that cells treated with nicotinamide can increase autophagy-associated LC3 conversion and double-membrane formation during GAS infection. Confocal microscopy images further showed that more GAS-containing vacuoles were colocalized with lysosome under nicotinamide-supplemented conditions than without nicotinamide treatment. In contrast to GAS, supplementation with exogenous nicotinamide did not effectively inhibit the growth of MRSA or S. Typhimurium in endothelial cells. These results indicate that intracellular NAD+ homeostasis is crucial for controlling intracellular GAS infection in endothelial cells. In addition, nicotinamide may be a potential new therapeutic agent to overcome persistent infections of GAS.
Collapse
Affiliation(s)
- Cheng-Lu Hsieh
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shu-Ying Hsieh
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hsuan-Min Huang
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shiou-Ling Lu
- Center for Frontier Oral Science, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Hiroko Omori
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Po-Xing Zheng
- Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yen-Ning Ho
- Department of Biotechnology and Laboratory Science in Medicine, School of Biomedical Science and Engineering, National Yang-Ming University, Taipei, Taiwan
| | - Yi-Lin Cheng
- Department of Biotechnology and Laboratory Science in Medicine, School of Biomedical Science and Engineering, National Yang-Ming University, Taipei, Taiwan
| | - Yee-Shin Lin
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chuan Chiang-Ni
- Department of Microbiology & Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Pei-Jane Tsai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shu-Ying Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ching-Chuan Liu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Pediatrics, College of Medicine, National Cheng Kung University and Hospital, Tainan, Taiwan
| | - Takeshi Noda
- Center for Frontier Oral Science, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Jiunn-Jong Wu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Biotechnology and Laboratory Science in Medicine, School of Biomedical Science and Engineering, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
40
|
Maiese K. Nicotinamide: Oversight of Metabolic Dysfunction Through SIRT1, mTOR, and Clock Genes. Curr Neurovasc Res 2020; 17:765-783. [PMID: 33183203 PMCID: PMC7914159 DOI: 10.2174/1567202617999201111195232] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/24/2020] [Accepted: 10/27/2020] [Indexed: 12/13/2022]
Abstract
Metabolic disorders that include diabetes mellitus present significant challenges for maintaining the welfare of the global population. Metabolic diseases impact all systems of the body and despite current therapies that offer some protection through tight serum glucose control, ultimately such treatments cannot block the progression of disability and death realized with metabolic disorders. As a result, novel therapeutic avenues are critical for further development to address these concerns. An innovative strategy involves the vitamin nicotinamide and the pathways associated with the silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1), the mechanistic target of rapamycin (mTOR), mTOR Complex 1 (mTORC1), mTOR Complex 2 (mTORC2), AMP activated protein kinase (AMPK), and clock genes. Nicotinamide maintains an intimate relationship with these pathways to oversee metabolic disease and improve glucose utilization, limit mitochondrial dysfunction, block oxidative stress, potentially function as antiviral therapy, and foster cellular survival through mechanisms involving autophagy. However, the pathways of nicotinamide, SIRT1, mTOR, AMPK, and clock genes are complex and involve feedback pathways as well as trophic factors such as erythropoietin that require a careful balance to ensure metabolic homeostasis. Future work is warranted to gain additional insight into these vital pathways that can oversee both normal metabolic physiology and metabolic disease.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, New York, New York 10022
| |
Collapse
|
41
|
Guarino M, Dufour JF. Nicotinamide and NAFLD: Is There Nothing New Under the Sun? Metabolites 2019; 9:E180. [PMID: 31510030 PMCID: PMC6780119 DOI: 10.3390/metabo9090180] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 09/05/2019] [Accepted: 09/06/2019] [Indexed: 12/18/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD) has a critical role in cellular metabolism and energy homeostasis. Its importance has been established early with the discovery of NAD's therapeutic role for pellagra. This review addresses some of the recent findings on NAD physiopathology and their effects on nonalcoholic fatty liver disease (NAFLD) pathogenesis, which need to be considered in the search for a better therapeutic approach. Reduced NAD concentrations contribute to the dysmetabolic imbalance and consequently to the pathogenesis of NAFLD. In this perspective, the dietary supplementation or the pharmacological modulation of NAD levels appear to be an attractive strategy. These reviewed studies open the doors to growing interest in NAD metabolism for NAFLD diagnosis, prevention, and treatment. Future rigorous clinical studies in humans will be necessary to validate these preliminary but promising results.
Collapse
Affiliation(s)
- Maria Guarino
- Hepatology, Department for BioMedical Research, University of Bern, 3008 Bern, Switzerland.
- Gastroenterology, Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy.
| | - Jean-François Dufour
- Hepatology, Department for BioMedical Research, University of Bern, 3008 Bern, Switzerland.
- University Clinic of Visceral Surgery and Medicine, Inselspital Bern, 3008 Bern, Switzerland.
| |
Collapse
|
42
|
Yang L, Wei J, Sheng F, Li P. Attenuation of Palmitic Acid-Induced Lipotoxicity by Chlorogenic Acid through Activation of SIRT1 in Hepatocytes. Mol Nutr Food Res 2019; 63:e1801432. [PMID: 31168914 DOI: 10.1002/mnfr.201801432] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 03/28/2019] [Indexed: 12/22/2022]
Abstract
SCOPE Saturated free fatty acids (FFAs) induce hepatocyte lipotoxicity, wherein oxidative stress-associated mitochondrial dysfunction is mechanistically involved. Chlorogenic acid (CGA), a potent antioxidant and anti-inflammatory compound, protects against high-fat-diet-induced oxidative stress and mitochondrial dysfunction in liver. This study investigates whether CGA protects against FFA-induced hepatocyte lipotoxicity via the regulation of mitochondrial fission/fusion and elucidates its underlying mechanisms. METHODS AND RESULTS AML12 cell, a non-transformed hepatocyte cell line, is treated with palmitate. Here, it is shown that CGA prevents palmitate-induced lipotoxicity by activation of SIRT1 regulated mitochondrial morphology. CGA treatment mitigates oxidative stress and mitochondrial dysfunction, as evidenced by a decrease in reactive oxygen species (ROS) production, and an increase in mitochondrial mass and mitochondrial membrane potential. CGA also significantly decreases Bax expression and thereby reduces mitochondria-mediated caspase-dependent apoptosis. Mechanistically, CGA attenuates ROS-induced mitochondrial fragmentation by inhibiting dynamin-related protein 1 (Drp1) and enhancing Mfn2 expression. In contrast, the inhibitory effects of CGA on the generation of mitochondrial ROS and Drp1 are blocked by siRNA knockdown of SIRT1. CONCLUSION Collectively, these findings show that supplementation with CGA protects hepatocytes from FFA-induced lipotoxicity through activation of SIRT1, which reverses the oxidative stress and dysfunction of mitochondrial biogenesis directly.
Collapse
Affiliation(s)
- Lele Yang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, 999078, China
| | - Jinchao Wei
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, 999078, China
| | - Feiya Sheng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, 999078, China
| | - Peng Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, 999078, China
| |
Collapse
|
43
|
Méndez-Lara KA, Santos D, Farré N, Nan MN, Pallarès V, Pérez-Pérez A, Alonso N, Escolà-Gil JC, Blanco-Vaca F, Julve J. Vitamin B3 impairs reverse cholesterol transport in Apolipoprotein E-deficient mice. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS 2019; 31:251-260. [PMID: 31097214 DOI: 10.1016/j.arteri.2019.04.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 03/18/2019] [Accepted: 04/01/2019] [Indexed: 01/06/2023]
Abstract
INTRODUCTION High Density Lipoproteins (HDL) are dysfunctional in hypercholesterolemia patients. The hypothesis was tested that nicotinamide (NAM) administration will influence HDL metabolism and reverse cholesterol transport from macrophages to the liver and feces in vivo (m-RCT) in a murine model of hypercholesterolemia. METHODS Apolipoprotein E-deficient (KOE) mice were challenged with a high-fat diet for 4 weeks. The effect of different doses of NAM on cholesterol metabolism, and the ability of HDL to promote m-RCT was assessed. RESULTS The administration of NAM to KOE mice produced an increase (∼1.5-fold; P<0.05) in the plasma levels of cholesterol, which was mainly accounted for by the non-HDL fraction. NAM produced a [3H]-cholesterol plasma accumulation (∼1.5-fold) in the m-RCT setting. As revealed by kinetic analysis, the latter was mainly explained by an impaired clearance of circulating non-HDL (∼0.8-fold). The relative content of [3H]-tracer was lowered in the livers (∼0.6-fold) and feces (>0.5-fold) of NAM-treated mice. This finding was accompanied by a significant (or trend close to significance) up-regulation of the relative gene expression of Abcg5 and Abcg8 in the liver (Abcg5: 2.9-fold; P<0.05; Abcg8: 2.4-fold; P=0.06) and small intestine (Abcg5: 2.1-fold; P=0.15; Abcg8: 1.9-fold; P<0.05) of high-dose, NAM-treated mice. CONCLUSION The data from this study show that the administration of NAM to KOE mice impaired m-RCT in vivo. This finding was partly due to a defective hepatic clearance of plasma non-HDL.
Collapse
Affiliation(s)
- Karen Alejandra Méndez-Lara
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau i Institut d'Investigació Biomèdica de l'Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain; Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - David Santos
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain
| | - Núria Farré
- Servei de Bioquímica, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Madalina Nicoleta Nan
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain; Servei de Bioquímica, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Víctor Pallarès
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau i Institut d'Investigació Biomèdica de l'Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Antonio Pérez-Pérez
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain; Servei d'Endocrinologia, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Núria Alonso
- Servei d'Endocrinologia, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain
| | - Joan Carles Escolà-Gil
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau i Institut d'Investigació Biomèdica de l'Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain; Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Francisco Blanco-Vaca
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain; Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain; Servei de Bioquímica, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Josep Julve
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau i Institut d'Investigació Biomèdica de l'Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain; Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
44
|
Zhao S, Zhang L, Yang C, Li Z, Rong S. Procyanidins and Alzheimer’s Disease. Mol Neurobiol 2019; 56:5556-5567. [DOI: 10.1007/s12035-019-1469-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 01/07/2019] [Indexed: 02/07/2023]
|
45
|
Li G, Yang J, Yang C, Zhu M, Jin Y, McNutt MA, Yin Y. PTENα regulates mitophagy and maintains mitochondrial quality control. Autophagy 2018; 14:1742-1760. [PMID: 29969932 DOI: 10.1080/15548627.2018.1489477] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
PTEN plays an important role in tumor suppression, and PTEN family members are involved in multiple biological processes in various subcellular locations. Here we report that PTENα, the first identified PTEN isoform, regulates mitophagy through promotion of PARK2 recruitment to damaged mitochondria. We show that PTENα-deficient mice exhibit accumulation of cardiac mitochondria with structural and functional abnormalities, and PTENα-deficient mouse hearts are more susceptible to injury induced by isoprenaline and ischemia-reperfusion. Mitochondrial clearance by mitophagy is also impaired in PTENα-deficient cardiomyocytes. In addition, we found PTENα physically interacts with the E3 ubiquitin ligase PRKN, which is an important mediator of mitophagy. PTENα binds PRKN through the membrane binding helix in its N-terminus, and promotes PRKN mitochondrial translocation through enhancing PRKN self-association in a phosphatase-independent manner. Loss of PTENα compromises mitochondrial translocation of PRKN and resultant mitophagy following mitochondrial depolarization. We propose that PTENα functions as a mitochondrial quality controller that maintains mitochondrial function and cardiac homeostasis. ABBREVIATIONS BECN1 beclin 1; CCCP carbonyl cyanide m-chlorophenylhydrazone; FBXO7 F-box protein 7; FS fraction shortening; HSPA1L heat shock protein family A (Hsp70) member 1 like; HW: BW heart weight:body weight ratio; I-R ischemia-reperfusion; ISO isoprenaline; MAP1LC3/LC3 microtubule associated protein 1 light chain 3; MBH membrane binding helix; MFN1 mitofusin 1; MFN2 mitofusin 2; Nam nicotinamide; TMRM tetramethylrhodamine ethyl ester; WGA wheat germ agglutinin.
Collapse
Affiliation(s)
- Guoliang Li
- a Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking-Tsinghua Center for Life Sciences , Peking University Health Science Center , Beijing , China
| | - Jingyi Yang
- b Institute of Systems Biomedicine, Department of Radiation Medicine, School of Basic Medical Sciences , Peking University Health Science Center , Beijing , China
| | - Chunyuan Yang
- a Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking-Tsinghua Center for Life Sciences , Peking University Health Science Center , Beijing , China
| | - Minglu Zhu
- a Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking-Tsinghua Center for Life Sciences , Peking University Health Science Center , Beijing , China
| | - Yan Jin
- a Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking-Tsinghua Center for Life Sciences , Peking University Health Science Center , Beijing , China
| | - Michael A McNutt
- a Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking-Tsinghua Center for Life Sciences , Peking University Health Science Center , Beijing , China
| | - Yuxin Yin
- a Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking-Tsinghua Center for Life Sciences , Peking University Health Science Center , Beijing , China
| |
Collapse
|
46
|
Abstract
Obesity poses a severe threat to human health, including the increased prevalence of hypertension, insulin resistance, diabetes mellitus, cancer, inflammation, sleep apnoea and other chronic diseases. Current therapies focus mainly on suppressing caloric intake, but the efficacy of this approach remains poor. A better understanding of the pathophysiology of obesity will be essential for the management of obesity and its complications. Knowledge gained over the past three decades regarding the aetiological mechanisms underpinning obesity has provided a framework that emphasizes energy imbalance and neurohormonal dysregulation, which are tightly regulated by autophagy. Accordingly, there is an emerging interest in the role of autophagy, a conserved homeostatic process for cellular quality control through the disposal and recycling of cellular components, in the maintenance of cellular homeostasis and organ function by selectively ridding cells of potentially toxic proteins, lipids and organelles. Indeed, defects in autophagy homeostasis are implicated in metabolic disorders, including obesity, insulin resistance, diabetes mellitus and atherosclerosis. In this Review, the alterations in autophagy that occur in response to nutrient stress, and how these changes alter the course of obesogenesis and obesity-related complications, are discussed. The potential of pharmacological modulation of autophagy for the management of obesity is also addressed.
Collapse
Affiliation(s)
- Yingmei Zhang
- Department of Cardiology, Fudan University Zhongshan Hospital, Shanghai, China.
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY, USA.
| | - James R Sowers
- Diabetes and Cardiovascular Research Center, University of Missouri-Columbia School of Medicine, Columbia, MO, USA
| | - Jun Ren
- Department of Cardiology, Fudan University Zhongshan Hospital, Shanghai, China.
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY, USA.
| |
Collapse
|
47
|
Wu WKK, Zhang L, Chan MTV. Autophagy, NAFLD and NAFLD-Related HCC. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1061:127-138. [PMID: 29956211 DOI: 10.1007/978-981-10-8684-7_10] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) will become a dominant cause of hepatocellular carcinoma (HCC) in the coming decade. Whereas the exact molecular mechanisms underlying the progression from simple steatosis, through steatohepatitis, to HCC remains largely unclear, emerging evidence has supported a central role of defective autophagy in the pathogenesis of NAFLD and its complications. Autophagy not only regulates lipid metabolism and insulin resistance, but also protects hepatocytes from injury and cell death. Nevertheless, in inflammation and tumorigenesis, the role of autophagy is more paradoxical. In NAFLD, defective hepatic autophagy occurs at multiple levels through numerous mechanisms and is causally linked to NAFLD-related HCC. In this chapter, we summarize the regulation and function of autophagy in NAFLD and highlight recent identification of potential pharmacological agents for restoring autophagic flux in NAFLD.
Collapse
Affiliation(s)
- William K K Wu
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong, Hong Kong. .,State Key Laboratory of Digestive Diseases, Department of Medicine & Therapeutics and LKS Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong.
| | - Lin Zhang
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong, Hong Kong.,State Key Laboratory of Digestive Diseases, Department of Medicine & Therapeutics and LKS Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Matthew T V Chan
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|