1
|
Yin Y, Guo H, Chen J, Yang Y, Li X, Zhang X. Bioactive Polysaccharides From Cremastra Appendiculata Attenuate Lipid Accumulation In Vitro Model of Non-Alcoholic Fatty Liver Disease. Chem Biodivers 2025:e202402772. [PMID: 39843986 DOI: 10.1002/cbdv.202402772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/18/2025] [Accepted: 01/22/2025] [Indexed: 01/24/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic disease with a rapidly growing incidence worldwide, presenting as an ever-increasing burden to the healthcare system. In this study, we explored that Cremastra appendiculata, a valuable traditional medicine in China, could alleviate lipid accumulation in HepG2 cells. C. appendiculata polysaccharide (CAP) was extracted from the pseudobulbs of C. appendiculata and oligosaccharide (oligoCAP) was obtained by enzymatical digestion of the CAP with endo-β-mannanase. Molecular weight of CAP and oligoCAP was determined to be 46 415 and 933 Da. Oleic acid (OA) induced steatosis in HepG2 cells was adopted as an in vitro NAFLD model. CAP and oligoCAP could reduce the OA-induced lipid accumulation and significantly reduce the total cholesterol and triglyceride content in the HepG2 cells. CAP and oligoCAP possessed lipid-lowering and anti-inflammatory activities especially by enhancing lipolysis and suppressing lipid synthesis shown by quantitative polymerase chain reaction results. Transcriptome analysis showed that CAP and oligoCAP mainly played anti-NAFLD roles in the pathways of lipid metabolism and necroptosis. Moreover, in vivo biodistribution results showed that both CAP and oligoCAP were mostly distributed in the small intestine and stomach tissues 24 h after administration, which may be related to the structural and molecular weight characterization of the polysaccharides.
Collapse
Affiliation(s)
- Yuan Yin
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Haiyang Guo
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Jiang Chen
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Yang Yang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Xiaojun Li
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| | - Xue Zhang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| |
Collapse
|
2
|
Chen S, Huang W, Huang T, Fang C, Zhao K, Zhang Y, Li H, Wu C. Highly sensitive near-infrared fluorescent probe for monitoring peroxynitrite in nonalcoholic fatty liver disease: Toward early diagnosis and therapeutic evaluation. Talanta 2025; 281:126865. [PMID: 39265422 DOI: 10.1016/j.talanta.2024.126865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/31/2024] [Accepted: 09/08/2024] [Indexed: 09/14/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) poses a significant global health concern, necessitating precise diagnostic tools and effective treatment strategies. Peroxynitrite (ONOO-), a reactive oxygen species, plays a pivotal role in NAFLD pathogenesis, highlighting its potential as a biomarker for disease diagnosis and therapeutic evaluation. This study reports on the development of a near-infrared (NIR) fluorescent probe, designated DRP-O, for the selective detection of ONOO- with high sensitivity and photostability. DRP-O exhibits rapid response kinetics (within 2 min) and an impressive detection limit of 2.3 nM, enabling real-time monitoring of ONOO- dynamics in living cells. Notably, DRP-O demonstrates excellent photostability under continuous laser irradiation, ensuring reliable long-term monitoring in complex biological systems. We apply DRP-O to visualize endogenous ONOO- in living cells, demonstrating its potential for diagnosing and monitoring NAFLD-related oxidative stress. Furthermore, DRP-O effectively evaluates the efficacy of therapeutic drugs in NAFLD cell models, underscoring its potential utility in drug screening studies. Moreover, we confirm DRP-O to enable selective identification of fatty liver tissues in a mouse model of NAFLD, indicating its potential for the early diagnosis of NAFLD. Collectively, DRP-O represents a valuable tool for studying ONOO- dynamics, evaluating drug efficacy, and diagnosing NAFLD, offering insights into novel therapeutic strategies for this prevalent liver disorder.
Collapse
Affiliation(s)
- Shiying Chen
- School of Materials and Chemical Engineering, Ningbo University of Technology, Ningbo, 315211, PR China; Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, PR China
| | - Wei Huang
- School of Materials and Chemical Engineering, Ningbo University of Technology, Ningbo, 315211, PR China; Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, PR China
| | - Ting Huang
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, PR China
| | - Cong Fang
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, PR China
| | - Kuicheng Zhao
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, PR China
| | - Youyu Zhang
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, PR China
| | - Haitao Li
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, PR China
| | - Cuiyan Wu
- School of Materials and Chemical Engineering, Ningbo University of Technology, Ningbo, 315211, PR China; Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, PR China.
| |
Collapse
|
3
|
Cui X, Li H, Li L, Xie C, Gao J, Chen Y, Zhang H, Hao W, Fu J, Guo H. Rodent model of metabolic dysfunction-associated fatty liver disease: a systematic review. J Gastroenterol Hepatol 2025; 40:48-66. [PMID: 39322221 PMCID: PMC11771679 DOI: 10.1111/jgh.16749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/30/2024] [Accepted: 09/11/2024] [Indexed: 09/27/2024]
Abstract
Although significant progress has been made in developing preclinical models for metabolic dysfunction-associated steatotic liver disease (MASLD), few have encapsulated the essential biological and clinical outcome elements reflective of the human condition. We conducted a comprehensive literature review of English-language original research articles published from 1990 to 2023, sourced from PubMed, Embase, and Web of Science, aiming to collate studies that provided a comparative analysis of physiological, metabolic, and hepatic histological characteristics between MASLD models and control groups. The establishment of a robust metabolic dysfunction-associated steatotic liver rodent model hinges on various factors, including animal species and strains, sex, induction agents and methodologies, and the duration of induction. Through this review, we aim to guide researchers in selecting suitable induction methods and animal species for constructing preclinical models aligned with their specific research objectives and laboratory conditions. Future studies should strive to develop simple, reliable, and reproducible models, considering the model's sensitivity to factors such as light-dark cycles, housing conditions, and environmental temperature. Additionally, the potential of diverse in vitro models, including 3D models and liver organ technology, warrants further exploration as valuable tools for unraveling the cellular mechanisms underlying fatty liver disease.
Collapse
Affiliation(s)
- Xiao‐Shan Cui
- Institute of Basic Medical Sciences, Xiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Hong‐Zheng Li
- Guang'an men HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Liang Li
- Institute of Basic Medical Sciences, Xiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Cheng‐Zhi Xie
- Institute of Basic Medical Sciences, Xiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Jia‐Ming Gao
- Institute of Basic Medical Sciences, Xiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Yuan‐Yuan Chen
- Institute of Basic Medical Sciences, Xiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Hui‐Yu Zhang
- Institute of Basic Medical Sciences, Xiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Wei Hao
- Institute of Basic Medical Sciences, Xiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Jian‐Hua Fu
- Institute of Basic Medical Sciences, Xiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Hao Guo
- Safety Laboratory, Xiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| |
Collapse
|
4
|
Wei Y, Zhang Y, Zhan B, Wang Y, Cheng J, Yu H, Lv M, Zhang Y, Zhai Y, Guan Y, Feng H. Asiaticoside alleviated NAFLD by activating Nrf2 and inhibiting the NF-κB pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156317. [PMID: 39667137 DOI: 10.1016/j.phymed.2024.156317] [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: 10/02/2023] [Revised: 09/18/2024] [Accepted: 12/04/2024] [Indexed: 12/14/2024]
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is a globally prevalent clinical problem of chronic liver disorder whose main pathogenic mechanisms are hepatic lipid accumulation, oxidative stress, and inflammatory reactions. Asiaticoside (Asi) is a compound derived from Centella asiatica, possesses antioxidant, antiphlogistic, antifibrotic, as well as wound healing properties. PURPOSE The aim of this study was to investigate the effects of Asi on NAFLD and its potential mechanisms. STUDY DESIGN Two versions of experimental models were constructed using free fatty acids (FFAs)-stimulated HepG2 cells along with high-fat diet (HFD)-incited NAFLD in mice. METHODS The pivotal action of Nrf2 was then explored using Ml-385 or Nrf2-/- mice. RESULTS The results indicated that Asi activated the Nrf2 to alleviate oxidative stress, inhibited the NF-κB to reduce the inflammatory response, and notably decreased lipid droplets and alleviated steatosis. CONCLUSION In conclusion, Asi demonstrates a potential to activate Nrf2 as well as inhibit the NF-κB pathway, there alleviate NAFLD.
Collapse
Affiliation(s)
- Yunfei Wei
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, PR China
| | - Yibo Zhang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, PR China
| | - Baihe Zhan
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, PR China
| | - Yajie Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, PR China
| | - Jiaqi Cheng
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, PR China
| | - Hao Yu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, PR China
| | - Mengfan Lv
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, PR China
| | - Yanmin Zhang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, PR China
| | - Yaxin Zhai
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, PR China
| | - Yuan Guan
- College of Animal Science, Jilin University, 5333 Xi'an Road, Changchun, Jilin 130062, PR China.
| | - Haihua Feng
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, PR China.
| |
Collapse
|
5
|
Niu QQ, Xi YT, Zhang CR, Li XY, Li CZ, Wang HD, Li P, Yin YL. Potential mechanism of perillaldehyde in the treatment of nonalcoholic fatty liver disease based on network pharmacology and molecular docking. Eur J Pharmacol 2024; 985:177092. [PMID: 39510336 DOI: 10.1016/j.ejphar.2024.177092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 10/08/2024] [Accepted: 10/31/2024] [Indexed: 11/15/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the most common chronic metabolic liver diseases worldwide. Perillaldehyde (4-propyl-1-en-2-ylcyclohexene-1-aldehyde, PA) is a terpenoid compound extracted from Perilla, which has effective pharmacological activities such as anti-inflammatory, antidepressant, and anticancer. This study aimed to explore the pharmacological effects of PA in intervening with NAFLD and reveal its potential mechanisms. Firstly, we identified the core targets of PA intervention therapy for NAFLD through network pharmacology and molecular docking techniques. After that, in vitro animal experiments such as H&E and Masson staining, immunofluorescence, immunohistochemistry, and Western blot were conducted to validate the results network effectively pharmacology predicted. Network pharmacology analysis suggested that PPAR-α may be the core target of PA intervention in NAFLD. H&E and Masson staining showed that after low-dose (50 mg/kg) PA administration, there was a noticeable improvement in fat deposition in the livers of NAFLD mice, and liver tissue fibrosis was alleviated. Immunohistochemical and immunofluorescence analysis showed that low dose (50 mg/kg) PA could reduce hepatocyte apoptosis, decrease the content of pro-apoptosis protein Bax, and increase the expression of anti-apoptosis protein Bcl-2 in NAFLD mice. Western blot results confirmed that low-dose (50 mg/kg) PA could increase the expression of PPAR-α and inhibit the expression of NF-κB in NAFLD mice. Our study indicated that PA could enhance the activity of PPAR-α and reduce the level of NF-κB in NAFLD mice, which may positively affect the prevention of NAFLD.
Collapse
Affiliation(s)
- Qian-Qian Niu
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China; Department of Toxicology, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang, 13200, Malaysia
| | - Yu-Ting Xi
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, 453003, China
| | - Chun-Rui Zhang
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, 453003, China
| | - Xi-Yue Li
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, 453003, China
| | - Cheng-Zhi Li
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, 453003, China
| | - Hui-Dan Wang
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, 453003, China
| | - Peng Li
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, 453003, China.
| | - Ya-Ling Yin
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
6
|
Suryaningtyas IT, Je JY. Therapeutic effects of blue mussel-derived peptides (PIISVYWK and FSVVPSPK) on non-alcoholic fatty liver disease by modulating lipid metabolism and inflammation in high-fat diet-induced mice. Tissue Cell 2024; 91:102630. [PMID: 39579737 DOI: 10.1016/j.tice.2024.102630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/13/2024] [Accepted: 11/18/2024] [Indexed: 11/25/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a progressive condition, advancing from simple hepatic lipid accumulation to inflammation, fibrosis, and increased risk of mortality. This study explores the therapeutic efficacy of bioactive peptides PIISVYWK (P1) and FSVVPSPK (P2) in ameliorating NAFLD in both oleic acid-treated HepG2 cells and high-fat diet (HFD)-induced mice. Our findings demonstrated that P1 and P2 significantly reduced hepatic fat deposition, enhanced lipolysis by promoting the release of free glycerol and free fatty acids, and suppressed key de novo lipogenesis-related proteins, including peroxisome proliferator-activated receptor γ (PPARγ), CCAAT-enhancer-binding protein α (C/EBPα), sterol regulatory element-binding protein 1 (SREBP-1), and fatty acid synthase (FAS). Furthermore, both peptides stimulated fatty acid oxidation via phosphorylation of AMP-activated protein kinase (AMPK) and hormone-sensitive lipase (HSL). Notably, reductions in body and liver weight, along with improved cholesterol profiles and liver function markers (alanine transaminase and aspartate aminotransferase), were observed in HFD mice. Additionally, P1 and P2 significantly attenuated the production of pro-inflammatory cytokines, such as tumor necrosis factor α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6) in both in vitro and in vivo models. Collectively, these results highlight the potent therapeutic potential of P1 and P2 in mitigating NAFLD progression, offering a promising intervention for this increasingly prevalent metabolic disorder.
Collapse
Affiliation(s)
- Indyaswan Tegar Suryaningtyas
- Department of Nutrition, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Food Technology and Processing, National Research and Innovation Agency, Yogyakarta 55861, Indonesia
| | - Jae-Young Je
- Major of Human Bioconvergence, Division of Smart Healthcare, Pukyong National University, Busan 48513, Republic of Korea.
| |
Collapse
|
7
|
Li X, Zhou L, Zheng Y, He T, Guo H, Li J, Zhang J. Establishment of a non-alcoholic fatty liver disease model by high fat diet in adult zebrafish. Animal Model Exp Med 2024; 7:904-913. [PMID: 36942644 PMCID: PMC11680480 DOI: 10.1002/ame2.12309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 12/30/2022] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) has become the most common chronic liver disease in recent years, but the pathogenesis is not fully understood. Therefore, it is important to establish an effective animal model for studying NAFLD. METHODS Adult zebrafish were fed a normal diet or a high-fat diet combined with egg yolk powder for 30 days. Body mass index (BMI) was measured to determine overall obesity. Serum lipids were measured using triglyceride (TG) and total cholesterol (TC) kits. Liver lipid deposition was detected by Oil Red O staining. Liver injury was assessed by measuring glutathione aminotransferase (AST) and glutamic acid aminotransferase (ALT) levels. Reactive oxygen species (ROS) and malondialdehyde (MDA) were used to evaluate oxidative damage. The level of inflammation was assessed by qRT-PCR for pro-inflammatory factors. H&E staining was used for pathological histology. Caspase-3 immunofluorescence measured apoptosis. Physiological disruption was assessed via RNA-seq analysis of genes at the transcriptional level and validated by qRT-PCR. RESULTS The high-fat diet led to significant obesity in zebrafish, with elevated BMI, hepatic TC, and TG. Severe lipid deposition in the liver was observed by ORO and H&E staining, accompanied by massive steatosis and ballooning. Serum AST and ALT levels were elevated, and significant liver damage was observed. The antioxidant system in the body was severely imbalanced. Hepatocytes showed massive apoptosis. RNA-seq results indicated that several physiological processes, including endoplasmic reticulum stress, and glucolipid metabolism, were disrupted. CONCLUSION Additional feeding of egg yolk powder to adult zebrafish for 30 consecutive days can mimic the pathology of human nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
- Xiang Li
- Department of Nutrition, School of Public HealthGuangdong Medical UniversityZhanjiangChina
- Zhanjiang Key Laboratory of Zebrafish Model for Development and DiseaseAffiliated Hospital of Guangdong Medical UniversityZhanjiangChina
| | - Lei Zhou
- Zhanjiang Key Laboratory of Zebrafish Model for Development and DiseaseAffiliated Hospital of Guangdong Medical UniversityZhanjiangChina
| | - Yuying Zheng
- Zhanjiang Key Laboratory of Zebrafish Model for Development and DiseaseAffiliated Hospital of Guangdong Medical UniversityZhanjiangChina
| | - Taiping He
- Department of Nutrition, School of Public HealthGuangdong Medical UniversityZhanjiangChina
| | - Honghui Guo
- Department of Nutrition, School of Public HealthGuangdong Medical UniversityZhanjiangChina
| | - Jiangbin Li
- School of Medical TechnologyGuangdong Medical UniversityDongguanChina
| | - Jingjing Zhang
- Zhanjiang Key Laboratory of Zebrafish Model for Development and DiseaseAffiliated Hospital of Guangdong Medical UniversityZhanjiangChina
| |
Collapse
|
8
|
Lei Y, Ma XL, Liu T, Wang MJ, Kang JS, Yang J, Mi N. Lactucin ameliorates FFA-induced steatosis in HepG2 cells by modulating mitochondrial homeostasis through the SIRT1/PGC-1α signaling axis. Heliyon 2024; 10:e39890. [PMID: 39524853 PMCID: PMC11550614 DOI: 10.1016/j.heliyon.2024.e39890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 10/25/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Nonalcoholic fatty liver disease is a complex disease involving abnormal liver metabolism. Its strong association with metabolic dysfunction has led to a change in nomenclature to metabolism dysfunction-associated fatty liver disease (MAFLD). MAFLD pathogenesis involves abnormal accumulation of hepatic lipids that lead to the production of excess free fatty acids (FFAs), which in turn cause an imbalance in hepatic mitochondrial function. Lactucin, a natural compound extracted from Cichorium glandulosum Boiss. et Huet, regulates liver metabolism and protects the liver. However, the potential mechanisms underlying the lactucin-mediated effects in MAFLD require further investigation. In the present study, HepG2 cells were treated with FFAs to establish an in vitro model of MAFLD. Parameters related to lipid accumulation and mitochondrial function, including triglycerides (TG), oil red O-stained lipid droplets, reactive oxygen species (ROS), mitochondrial membrane potential (JC-1), adenine triphosphate (ATP), and complex III were analysed. Morphology of the mitochondria were evaluated by transmission electron microscopy. Furthermore, key proteins in the sirtuin 1 (SIRT1)/peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α) signalling axis and mitochondrial quality control were analysed. The SIRT1 inhibitor EX-527 was used to verify the key role of the SIRT1 signalling pathway. Western blotting showed that lactucin upregulated the expression of SIRT-1, PGC-1α, Nrf1, Tfam, Mfn2, and Opa1, and promoted mitochondrial biosynthesis and kinetics. The results suggest that lactucin restores mitochondrial dynamic homeostasis by upregulating the SIRT1/PGC-1α signalling axis, thereby reducing FFA-induced lipid accumulation in HepG2 cells.
Collapse
Affiliation(s)
- Yi Lei
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830011, China
| | - Xiao-li Ma
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830011, China
- Key Laboratory of Active Components of Xinjiang Natural Medicine and Drug Release Technology, Urumqi, 830000, China
| | - Tong Liu
- Basic Medical College, Xinjiang Medical University, Urumqi, 830011, China
| | - Meng-jiao Wang
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830011, China
| | - Jin-sen Kang
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830011, China
- Key Laboratory of Active Components of Xinjiang Natural Medicine and Drug Release Technology, Urumqi, 830000, China
| | - Jian Yang
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830011, China
- Key Laboratory of Active Components of Xinjiang Natural Medicine and Drug Release Technology, Urumqi, 830000, China
| | - Na Mi
- Key Laboratory of Active Components of Xinjiang Natural Medicine and Drug Release Technology, Urumqi, 830000, China
- Clinical Medicine Research Institute, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| |
Collapse
|
9
|
Dadkhah Nikroo N, Jafarinejad H, Yousefi Z, Abdolvahabi Z, Malek M, Mortazavi P, Pazouki A, Mokhber S, Nourbakhsh M. Elevated mir-141 in obesity: Insights into the interplay with sirtuin 1 and non-alcoholic fatty liver disease. Obes Sci Pract 2024; 10:e70007. [PMID: 39345780 PMCID: PMC11427942 DOI: 10.1002/osp4.70007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 08/21/2024] [Accepted: 08/25/2024] [Indexed: 10/01/2024] Open
Abstract
Background Changes in gene expression related to obesity are linked to microRNAs, such as miR-141, which play a crucial role in metabolic homeostasis. Sirtuin 1 (SIRT1), an enzyme that plays a crucial role in regulating various cellular functions and metabolism, is implicated in obesity and the ensuing non-alcoholic fatty liver disease (NAFLD). The aim of this research was to evaluate the levels of miR-141 and its relationship with SIRT1 and NAFLD. Methods A group of 100 adults (50 with obesity and 50 with normal-weight) were selected and underwent complete clinical evaluation and anthropometric measurements. Biochemical parameters were assessed in blood serum, and the levels of miR-141 in plasma were measured by real-time PCR. The expression of the SIRT1 gene was also evaluated in the peripheral blood mononuclear cells using Real-time PCR. The ELISA technique was used to determine insulin levels. Liver steatosis was assessed by ultrasound. Results The results showed that levels of miR-141 were significantly increased in participants with obesity compared with the control group. Conversely, the expression of the SIRT1 gene in individuals with obesity was lower than that in control participants. A strong negative correlation was observed between miR-141 and SIRT1 and a strong positive association was observed between miR-141 and metabolic parameters. Furthermore, participants with fatty liver had significantly elevated levels of miR-141 gene expression and lower expression of SIRT1 gene, compared to those without fatty liver. Conclusion elevated levels of miR-141 in individuals with obesity might be a contributing factor in the repression of SIRT1 in obesity and its consequences, including NAFLD. Therefore, miR-141 might serve as a suitable diagnostic and therapeutic target in obesity and NAFLD.
Collapse
Affiliation(s)
- Nikta Dadkhah Nikroo
- Finetech in Medicine Research CenterIran University of Medical SciencesTehranIran
| | - Habib Jafarinejad
- Cancer Research Center and Department of ImmunologySchool of MedicineSemnan University of Medical SciencesSemnanIran
- Legal Medicine Research CenterLegal Medicine OrganizationTehranIran
| | - Zeynab Yousefi
- Department of Clinical BiochemistryFaculty of Medical ScienceTarbiat Modares UniversityTehranIran
| | - Zohreh Abdolvahabi
- Cellular and Molecular Research CentreResearch Institute for Prevention of Non‐Communicable DiseasesQazvin University of Medical SciencesQazvinIran
| | - Mojtaba Malek
- Research Center for Prevention of Cardiovascular DiseaseInstitute of Endocrinology and MetabolismIran University of Medical SciencesTehranIran
| | - Pejman Mortazavi
- Department of PathobiologyFaculty of Veterinary MedicineScience and Research BranchIslamic Azad UniversityTehranIran
| | - Abdolreza Pazouki
- Minimally Invasive Surgery Research CenterIran University of Medical SciencesTehranIran
| | - Somayeh Mokhber
- Minimally Invasive Surgery Research CenterIran University of Medical SciencesTehranIran
| | - Mitra Nourbakhsh
- Finetech in Medicine Research CenterIran University of Medical SciencesTehranIran
- Department of BiochemistrySchool of MedicineIran University of Medical SciencesTehranIran
| |
Collapse
|
10
|
Zou J, Gu Q, Gu D. Association between phthalates exposure and non-alcoholic fatty liver disease under different diagnostic criteria: a cross-sectional study based on NHANES 2017 to 2018. Front Public Health 2024; 12:1407976. [PMID: 39386944 PMCID: PMC11462993 DOI: 10.3389/fpubh.2024.1407976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 09/09/2024] [Indexed: 10/12/2024] Open
Abstract
Purpose Non-alcoholic fatty liver disease (NAFLD) is the most common liver disease. Phthalates have been suggested to influence the development of NAFLD due to their endocrine-disrupting properties, but studies based on nationally representative populations are insufficient, and existing studies seem to have reached conflicting conclusions. Due to changes in legislation, the use of traditional phthalates has gradually decreased, and the phthalates substitutes is getting more attention. This study aims to delve deeper into how the choice of diagnostic approach influences observed correlations and concern about more alternatives of phthalates, thereby offering more precise references for the prevention and treatment of NAFLD. Methods A cohort of 641 participants, sourced from the National Health and Nutrition Examination Survey (NHANES) 2017-2018 database, was evaluated for NAFLD using three diagnostic methods: the Hepatic Steatosis Index (HSI), the US Fatty Liver Indicator (US.FLI), and Vibration Controlled Transient Elastography (VCTE). The urinary metabolite concentrations of Di-2-ethylhexyl phthalate (DEHP), Di-isodecyl phthalate (DIDP), Di-isononyl phthalate (DINP), Di-n-butyl phthalate (DnBP), Di-isobutyl phthalate (DIBP), Di-ethyl phthalate (DEP) and Di-n-octyl phthalate (DnOP) were detected. The association between NAFLD and urinary phthalate metabolites was evaluated through univariate and multivariate logistic regression analyses, considering different concentration gradients of urinary phthalates. Results Univariate logistic regression analysis found significant correlations between NAFLD and specific urinary phthalate metabolites, such as Mono-(2-ethyl-5-oxohexyl) phthalate (MEOHP), Mono-(2-ethyl-5-hydroxyhexyl) phthalate (MEHHP), Mono-2-ethyl-5-carboxypentyl phthalate (MECPP), and Mono-(carboxyisoctyl) phthalate (MCiOP), across different diagnostic criteria. In a multivariate logistic regression analysis adjusting only for demographic data, MEOHP (OR = 3.26, 95% CI = 1.19-8.94, p = 0.029), MEHHP (OR = 3.98, 95% CI = 1.43-11.1, p = 0.016), MECPP (OR = 3.52, 95% CI = 1.01-12.2, p = 0.049), and MCiOP (OR = 4.55, 95% CI = 1.93-10.7, p = 0.005) were positively related to NAFLD defined by HSI and VCTE. The correlation strength varied with the concentration of phthalates, indicating a potential dose-response relationship. Adjusting for all covariates in multivariate logistic regression, only MCiOP (OR = 4.22, 95% CI = 1.10-16.2, p = 0.044), as an oxidative metabolite of DINP, remained significantly associated with NAFLD under the VCTE criterion, suggesting its potential role as a risk factor for NAFLD. Conclusion This research highlights a significant association between DINP and NAFLD. These findings underscore the need for further investigation into the role of the phthalates substitutes in the pathogenesis of NAFLD and the importance of considering different diagnostic criteria in research.
Collapse
Affiliation(s)
- Jiazhen Zou
- Department of Laboratory Medicine, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen Key Laboratory of Medical Laboratory and Molecular Diagnostics, Shenzhen, China
| | - Qingdan Gu
- Shenzhen Yantian District People’s Hospital (Group), Southern University of Science and Technology Yantian Hospital, Shenzhen, China
| | - Dayong Gu
- Department of Laboratory Medicine, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen Key Laboratory of Medical Laboratory and Molecular Diagnostics, Shenzhen, China
| |
Collapse
|
11
|
Tripathi M, Gauthier K, Sandireddy R, Zhou J, Guptta P, Sakthivel S, Teo WW, Naing YT, Arul K, Tikno K, Park SH, Wu Y, Wang L, Bay BH, Sun L, Giguere V, Chow PKH, Ghosh S, McDonnell DP, Yen PM, Singh BK. Esrra regulates Rplp1-mediated translation of lysosome proteins suppressed in metabolic dysfunction-associated steatohepatitis and reversed by alternate day fasting. Mol Metab 2024; 87:101997. [PMID: 39032642 PMCID: PMC11327444 DOI: 10.1016/j.molmet.2024.101997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/03/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024] Open
Abstract
OBJECTIVE Currently, little is known about the mechanism(s) regulating global and specific protein translation during metabolic dysfunction-associated steatohepatitis (MASH; previously known as non-alcoholic steatohepatitis, NASH). METHODS Unbiased label-free quantitative proteome, puromycin-labelling and polysome profiling were used to understand protein translation activity in vitro and in vivo. RESULTS We observed a global decrease in protein translation during lipotoxicity in human primary hepatocytes, mouse hepatic AML12 cells, and livers from a dietary mouse model of MASH. Interestingly, proteomic analysis showed that Rplp1, which regulates ribosome and translation pathways, was one of the most downregulated proteins. Moreover, decreased Esrra expression and binding to the Rplp1 promoter, diminished Rplp1 gene expression during lipotoxicity. This, in turn, reduced global protein translation and Esrra/Rplp1-dependent translation of lysosome (Lamp2, Ctsd) and autophagy (sqstm1, Map1lc3b) proteins. Of note, Esrra did not increase its binding to these gene promoters or their gene transcription, confirming its regulation of their translation during lipotoxicity. Notably, hepatic Esrra-Rplp1-dependent translation of lysosomal and autophagy proteins also was impaired in MASH patients and liver-specific Esrra knockout mice. Remarkably, alternate day fasting induced Esrra-Rplp1-dependent expression of lysosomal proteins, restored autophagy, and reduced lipotoxicity, inflammation, and fibrosis in hepatic cell culture and in vivo models of MASH. CONCLUSIONS Esrra regulation of Rplp1-mediated translation of lysosome/autolysosome proteins was downregulated during MASH. Alternate day fasting activated this novel pathway and improved MASH, suggesting that Esrra and Rplp1 may serve as therapeutic targets for MASH. Our findings also provided the first example of a nuclear hormone receptor, Esrra, to not only regulate transcription but also protein translation, via induction of Rplp1.
Collapse
Affiliation(s)
- Madhulika Tripathi
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Karine Gauthier
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS, Ecole Normale Supérieure de Lyon, 46 Allée d'Italie 69364 Lyon Cedex 07, France
| | - Reddemma Sandireddy
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Jin Zhou
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Priyanka Guptta
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Suganya Sakthivel
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Wei Wen Teo
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Yadanar Than Naing
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Kabilesh Arul
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Keziah Tikno
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Sung-Hee Park
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, C238A Levine Science Research Center, Durham, NC 27710, USA
| | - Yajun Wu
- Department of Anatomy, Yong Loo Lin School of Medicine, NUS 117594, Singapore
| | - Lijin Wang
- Centre for Computational Biology, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore; Pennington Biomedical Research Center, Laboratory of Bioinformatics and Computational Biology, Baton Rouge, LA 70808, USA
| | - Boon-Huat Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, NUS 117594, Singapore
| | - Lei Sun
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Vincent Giguere
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue West, Montreal, Québec H3A 1A3, Canada
| | - Pierce K H Chow
- Dept of Surgery, Singapore General Hospital and Dept. of Surgical Oncology, National Cancer Centre 169608, Singapore
| | - Sujoy Ghosh
- Centre for Computational Biology, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore; Pennington Biomedical Research Center, Laboratory of Bioinformatics and Computational Biology, Baton Rouge, LA 70808, USA
| | - Donald P McDonnell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, C238A Levine Science Research Center, Durham, NC 27710, USA
| | - Paul M Yen
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore; Duke Molecular Physiology Institute and Dept. of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Brijesh K Singh
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore.
| |
Collapse
|
12
|
Somabattini RA, Sherin S, Siva B, Chowdhury N, Nanjappan SK. Unravelling the complexities of non-alcoholic steatohepatitis: The role of metabolism, transporters, and herb-drug interactions. Life Sci 2024; 351:122806. [PMID: 38852799 DOI: 10.1016/j.lfs.2024.122806] [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: 03/27/2024] [Revised: 05/24/2024] [Accepted: 06/04/2024] [Indexed: 06/11/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a mainstream halting liver disease with high prevalence in North America, Europe, and other world regions. It is an advanced form of NAFLD caused by the amassing of fat in the liver and can progress to the more severe form known as non-alcoholic steatohepatitis (NASH). Until recently, there was no authorized pharmacotherapy reported for NASH, and to improve the patient's metabolic syndrome, the focus is mainly on lifestyle modification, weight loss, ensuring a healthy diet, and increased physical activity; however, the recent approval of Rezdiffra (Resmetirom) by the US FDA may change this narrative. As per the reported studies, there is an increased articulation of uptake and efflux transporters of the liver, including OATP and MRP, in NASH, leading to changes in the drug's pharmacokinetic properties. This increase leads to alterations in the pharmacokinetic properties of drugs. Furthermore, modifications in Cytochrome P450 (CYP) enzymes can have a significant impact on these properties. Xenobiotics are metabolized primarily in the liver and constitute liver enzymes and transporters. This review aims to delve into the role of metabolism, transport, and potential herb-drug interactions in the context of NASH.
Collapse
Affiliation(s)
- Ravi Adinarayan Somabattini
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, Chunilal Bhawan, 168, Maniktala Main Road, Kolkata 700054, West Bengal, India
| | - Sahla Sherin
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, Chunilal Bhawan, 168, Maniktala Main Road, Kolkata 700054, West Bengal, India
| | - Bhukya Siva
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, Chunilal Bhawan, 168, Maniktala Main Road, Kolkata 700054, West Bengal, India
| | - Neelanjan Chowdhury
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, Chunilal Bhawan, 168, Maniktala Main Road, Kolkata 700054, West Bengal, India
| | - Satheesh Kumar Nanjappan
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, Chunilal Bhawan, 168, Maniktala Main Road, Kolkata 700054, West Bengal, India.
| |
Collapse
|
13
|
Pei Y, He Y, Wang X, Xie C, Li L, Sun Q, Liu L, Shan S, Wang P, Liu T, Fan X, Cong M, Jia J. Tartaric acid ameliorates experimental non-alcoholic fatty liver disease by activating the AMP-activated protein kinase signaling pathway. Eur J Pharmacol 2024; 975:176668. [PMID: 38788791 DOI: 10.1016/j.ejphar.2024.176668] [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: 02/19/2024] [Revised: 05/01/2024] [Accepted: 05/22/2024] [Indexed: 05/26/2024]
Abstract
Tartaric acid (TA) has been shown beneficial effects on blood pressure and lipid levels. However, its effect on non-alcoholic fatty liver disease (NAFLD) remains unknown. This study aimed to investigate the role of TA in experimental NAFLD. Mice were fed a Western diet for 8 weeks, followed by administration of TA or a vehicle for an additional 12 weeks while continuing on the Western diet. Blood biochemistry including transaminases and glucose tolerance test and liver tissue RNA sequencing (RNA-seq), lipid content, and histology were investigated. The HepG2 cell line was used to explore the mechanism by which TA regulates lipid metabolism. We found that TA significantly improved weight gain, insulin resistance, hepatic steatosis, inflammation and fibrosis in Western diet-fed mice. By comparing gene expression differences, we found that TA affects pathways related to lipid metabolism, inflammatory response, and fibrosis. Furthermore, TA effectively reduced oleic acid-induced lipid accumulation in HepG2 cells and downregulated the genes associated with fatty acid synthesis, which were enriched in the AMP-activated protein kinase (AMPK) signaling pathway. TA also enhanced the phosphorylation of AMPK which could be reverted by the AMPK inhibitor Compound C in HepG2 cells. Our study suggests that TA improves experimental NAFLD by activating the AMPK signaling pathway. These findings indicate that TA may serve as a potential therapy for the human NAFLD.
Collapse
Affiliation(s)
- Yufeng Pei
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, National Clinical Research Center for Digestive Diseases, State Key Lab of Digestive Health, Beijing, China
| | - Yu He
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, National Clinical Research Center for Digestive Diseases, State Key Lab of Digestive Health, Beijing, China
| | - Xiaofan Wang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, National Clinical Research Center for Digestive Diseases, State Key Lab of Digestive Health, Beijing, China
| | - Chao Xie
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, National Clinical Research Center for Digestive Diseases, State Key Lab of Digestive Health, Beijing, China
| | - Li Li
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, National Clinical Research Center for Digestive Diseases, State Key Lab of Digestive Health, Beijing, China
| | - Qingyun Sun
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, National Clinical Research Center for Digestive Diseases, State Key Lab of Digestive Health, Beijing, China
| | - Lin Liu
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, National Clinical Research Center for Digestive Diseases, State Key Lab of Digestive Health, Beijing, China
| | - Shan Shan
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, National Clinical Research Center for Digestive Diseases, State Key Lab of Digestive Health, Beijing, China
| | - Ping Wang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, National Clinical Research Center for Digestive Diseases, State Key Lab of Digestive Health, Beijing, China
| | - Tianhui Liu
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, National Clinical Research Center for Digestive Diseases, State Key Lab of Digestive Health, Beijing, China
| | - Xu Fan
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, National Clinical Research Center for Digestive Diseases, State Key Lab of Digestive Health, Beijing, China
| | - Min Cong
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, National Clinical Research Center for Digestive Diseases, State Key Lab of Digestive Health, Beijing, China.
| | - Jidong Jia
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, National Clinical Research Center for Digestive Diseases, State Key Lab of Digestive Health, Beijing, China.
| |
Collapse
|
14
|
Tang J, Wang L, Shi M, Feng S, Zhang T, Han H. Study on the mechanism of Shuganzhi Tablet against nonalcoholic fatty liver disease and lipid regulation effects of its main substances in vitro. JOURNAL OF ETHNOPHARMACOLOGY 2023; 316:116780. [PMID: 37311504 DOI: 10.1016/j.jep.2023.116780] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/30/2023] [Accepted: 06/10/2023] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shuganzhi Tablet (SGZT) originates from a famous traditional Chinese herbal formula Chaihu Decoction which can be applied to treat liver diseases, however, the pharmacodynamic mechanism of SGZT needs to be evaluated. AIM OF THIS STUDY To study the mechanism of SGZT in the treatment of non-alcoholic fatty liver disease (NAFLD), and screen out its effective ingredients. MATERIALS AND METHODS In this study, firstly, the main components of SGZT were analyzed qualitatively. And a rat model of NAFLD was established by feeding high-fat diet. Serum biochemical indexes and liver pathological analysis were used to evaluate the pharmacodynamic effect of SGZT in the treatment of NAFLD. In order to explore the pharmacodynamic mechanism, proteomics and metabolomics analysis were used. Western blotting was used to verify the expression of important differential proteins. And L02 cells were treated with free fatty acids (FFA) and the main substances of SGZT to establish the cell model of NAFLD in vitro and to reveal the pharmacodynamic substance of SGZT. RESULTS Twelve components were detected in SGZT, and according to the results of serum biochemical indexes and liver pathological analysis, SGZT could effectively treat NAFLD. Combined with the results of bioinformatics analysis, we found that 133 differentially expressed proteins were reversed in liver samples of rats treated with SGZT. The important proteins in PPAR signaling pathway, steroid biosynthesis, cholesterol metabolism and fatty acid metabolism were mainly regulated to maintain cholesterol homeostasis and improve lipid metabolism. SGZT also affected various metabolites in rat liver, including eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA) and taurine. In addition, the main components contained in SGZT (hesperidin, polydatin, naringin, emodin, specnuezhenide, saikosaponin A) and a metabolite (resveratrol) could significantly reduce FFA-induced intracellular lipid accumulation. CONCLUSION SGZT effectively treated NAFLD, and PPAR-γ, Acsl4, Plin2 and Fads1 may be the main targets of SGZT. And Fads1-EPA/DHA-PPAR-γ may be the potential pharmacodynamic pathway. Cell experiments in vitro revealed that the main components of SGZT and their metabolites, such as hesperidin, polydatin, naringin, emodin, specnuezhenide, saikosaponin A and resveratrol may be the main components of its efficacy. Further research is needed to reveal and validate the pharmacodynamic mechanism.
Collapse
Affiliation(s)
- Jie Tang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China.
| | - Lixiang Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China.
| | - Mengge Shi
- Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China.
| | - Shuaixia Feng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China.
| | - Tong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China.
| | - Han Han
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China.
| |
Collapse
|
15
|
Al Jadani JM, Albadr NA, Alshammari GM, Almasri SA, Alfayez FF, Yahya MA. Esculeogenin A, a Glycan from Tomato, Alleviates Nonalcoholic Fatty Liver Disease in Rats through Hypolipidemic, Antioxidant, and Anti-Inflammatory Effects. Nutrients 2023; 15:4755. [PMID: 38004149 PMCID: PMC10675668 DOI: 10.3390/nu15224755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
This study examined the preventative effects of esculeogenin A (ESGA), a newly discovered glycan from tomato, on liver damage and hepatic steatosis in high-fat-diet (HFD)-fed male rats. The animals were divided into six groups (each of eight rats): a control group fed a normal diet, control + ESGA (200 mg/kg), HFD, and HFD + ESAG in 3 doses (50, 100, and 200 mg/kg). Feeding and treatments were conducted for 12 weeks. Treatment with ESGA did not affect gains in the body or fat weight nor increases in fasting glucose, insulin, and HOMA-IR or serum levels of free fatty acids (FFAs), tumor-necrosis factor-α, and interleukin-6 (IL-6). On the contrary, it significantly reduced the serum levels of gamma-glutamyl transpeptidase (GGT), aspartate aminotransferase (AST), alanine aminotransferase (ALT), total triglycerides (TGs), cholesterol (CHOL), and low-density lipoprotein cholesterol (LDL-c) in the HFD-fed rats. In addition, it improved the liver structure, attenuating the increase in fat vacuoles; reduced levels of TGs and CHOL, and the mRNA levels of SREBP1 and acetyl CoA carboxylase (ACC); and upregulated the mRNA levels of proliferator-activated receptor α (PPARα) and carnitine palmitoyltransferase I (CPT I) in HFD-fed rats. These effects were concomitant with increases in the mRNA, cytoplasmic, and nuclear levels of nuclear factor erythroid 2-related factor 2 (Nrf2), glutathione (GSH), superoxide dismutase (SOD), catalase (CAT), and heme oxygenase-1 (HO); a reduction in the nuclear activity of nuclear factor-kappa beta (NF-κB); and inhibition of the activity of nuclear factor kappa B kinase subunit beta (IKKβ). All of these effects were dose-dependent effects in which a normal liver structure and normal levels of all measured parameters were seen in HFD + ESGA (200 mg/kg)-treated rats. In conclusion, ESGA prevents NAFLD in HFD-fed rats by attenuating hyperlipidemia, hepatic steatosis, oxidative stress, and inflammation by acting locally on Nrf2, NF-κB, SREBP1, and PPARα transcription factors.
Collapse
Affiliation(s)
- Jwharah M. Al Jadani
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia; (J.M.A.J.); (G.M.A.); (S.A.A.); (M.A.Y.)
| | - Nawal A. Albadr
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia; (J.M.A.J.); (G.M.A.); (S.A.A.); (M.A.Y.)
| | - Ghedeir M. Alshammari
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia; (J.M.A.J.); (G.M.A.); (S.A.A.); (M.A.Y.)
| | - Soheir A. Almasri
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia; (J.M.A.J.); (G.M.A.); (S.A.A.); (M.A.Y.)
| | - Farah Fayez Alfayez
- Department of Medicine and Surgery, College of Medicine, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Mohammed Abdo Yahya
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia; (J.M.A.J.); (G.M.A.); (S.A.A.); (M.A.Y.)
| |
Collapse
|
16
|
Massemin A, Goehrig D, Flaman J, Jaber S, Griveau A, Djebali S, Marcos E, Payen L, Marvel J, Parent R, Adnot S, Bertolino P, Rieusset J, Tortereau A, Vindrieux D, Bernard D. Loss of Pla2r1 decreases cellular senescence and age-related alterations caused by aging and Western diets. Aging Cell 2023; 22:e13971. [PMID: 37667516 PMCID: PMC10652324 DOI: 10.1111/acel.13971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 06/21/2023] [Accepted: 08/04/2023] [Indexed: 09/06/2023] Open
Abstract
Cellular senescence is induced by many stresses including telomere shortening, DNA damage, oxidative, or metabolic stresses. Senescent cells are stably cell cycle arrested and they secrete many factors including cytokines and chemokines. Accumulation of senescent cells promotes many age-related alterations and diseases. In this study, we investigated the role of the pro-senescent phospholipase A2 receptor 1 (PLA2R1) in regulating some age-related alterations in old mice and in mice subjected to a Western diet, whereas aged wild-type mice displayed a decreased ability to regulate their glycemia during glucose and insulin tolerance tests, aged Pla2r1 knockout (KO) mice efficiently regulated their glycemia and displayed fewer signs of aging. Loss of Pla2r1 was also found protective against the deleterious effects of a Western diet. Moreover, these Pla2r1 KO mice were partially protected from diet-induced senescent cell accumulation, steatosis, and fibrosis. Together these results support that Pla2r1 drives several age-related alterations, especially in the liver, arising during aging or through a Western diet.
Collapse
Affiliation(s)
- Amélie Massemin
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon BérardUniversité de LyonLyonFrance
- Equipe Labellisée la Ligue Contre le CancerLyonFrance
| | - Delphine Goehrig
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon BérardUniversité de LyonLyonFrance
- Equipe Labellisée la Ligue Contre le CancerLyonFrance
| | - Jean‐Michel Flaman
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon BérardUniversité de LyonLyonFrance
- Equipe Labellisée la Ligue Contre le CancerLyonFrance
| | - Sara Jaber
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon BérardUniversité de LyonLyonFrance
| | - Audrey Griveau
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon BérardUniversité de LyonLyonFrance
| | - Sophia Djebali
- Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, École Normale Supérieure de LyonUniversité de Lyon, Université Claude Bernard Lyon 1LyonFrance
| | - Elisabeth Marcos
- INSERM U955, Département de Physiologie ‐ Explorations fonctionnelles, Hôpital Henri MondorAP‐HP, FHU SENECCréteilFrance
| | - Léa Payen
- Laboratoire de Biochimie et Biologie Moléculaire, Centre Hospitalier Lyon SudHospices Civils de LyonPierre BéniteFrance
| | - Jacqueline Marvel
- Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, École Normale Supérieure de LyonUniversité de Lyon, Université Claude Bernard Lyon 1LyonFrance
| | - Romain Parent
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon BérardUniversité de LyonLyonFrance
| | - Serge Adnot
- INSERM U955, Département de Physiologie ‐ Explorations fonctionnelles, Hôpital Henri MondorAP‐HP, FHU SENECCréteilFrance
| | - Philippe Bertolino
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon BérardUniversité de LyonLyonFrance
- Equipe Labellisée la Ligue Contre le CancerLyonFrance
| | - Jennifer Rieusset
- CarMeN Laboratory, UMR INSERM U1060/INRA U1397Lyon 1 UniversityPierre béniteFrance
| | - Antonin Tortereau
- VetAgro Sup, Interactions Cellules Environnement (ICE)Université de LyonMarcy l'EtoileFrance
| | - David Vindrieux
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon BérardUniversité de LyonLyonFrance
- Equipe Labellisée la Ligue Contre le CancerLyonFrance
| | - David Bernard
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon BérardUniversité de LyonLyonFrance
- Equipe Labellisée la Ligue Contre le CancerLyonFrance
| |
Collapse
|
17
|
Kim W, Li M, Jin H, Yang H, Türkez H, Uhlén M, Zhang C, Mardinoglu A. Characterization of an in vitro steatosis model simulating activated de novo lipogenesis in MAFLD patients. iScience 2023; 26:107727. [PMID: 37674987 PMCID: PMC10477067 DOI: 10.1016/j.isci.2023.107727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/18/2023] [Accepted: 08/21/2023] [Indexed: 09/08/2023] Open
Abstract
Activated de novo lipogenesis (DNL) is the critical pathway involved in the progression of metabolic-associated fatty liver disease (MAFLD). We present an in vitro steatosis model for MAFLD that induces steatosis through activated DNL. This model utilizes insulin and LXR receptor ligand T0901317, eliminating the need for fatty acid treatment. Significant increases in triglycerides (TAGs) and expression of DNL-related transcription factors were observed. Transcriptomic analysis revealed distinct gene expression profiles between the DNL and conventional oleic acid (OA)-induced steatosis model. DNL steatosis model exhibited elevated pathways related to glycolysis, cholesterol homeostasis, and bile acid metabolism, reflecting its clinical relevance to MAFLD. Moreover, C75 and JNK-IN-5A compounds effectively reduced TAG accumulation and steatosis-related protein expression in the DNL model, whereas they had no significant impact on TAG accumulation in the OA model. In conclusion, we introduce an ideal model for steatosis study, which could help in understanding the MAFLD mechanisms.
Collapse
Affiliation(s)
- Woonghee Kim
- Science for Life Laboratory, KTH – Royal Institute of Technology, Stockholm 17165, Sweden
| | - Mengzhen Li
- Science for Life Laboratory, KTH – Royal Institute of Technology, Stockholm 17165, Sweden
| | - Han Jin
- Science for Life Laboratory, KTH – Royal Institute of Technology, Stockholm 17165, Sweden
| | - Hong Yang
- Science for Life Laboratory, KTH – Royal Institute of Technology, Stockholm 17165, Sweden
| | - Hasan Türkez
- Department of Medical Biology, Faculty of Medicine, Atatürk University, Erzurum, Turkey
| | - Mathias Uhlén
- Science for Life Laboratory, KTH – Royal Institute of Technology, Stockholm 17165, Sweden
| | - Cheng Zhang
- Science for Life Laboratory, KTH – Royal Institute of Technology, Stockholm 17165, Sweden
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH – Royal Institute of Technology, Stockholm 17165, Sweden
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London SE1 9RT, UK
| |
Collapse
|
18
|
Zhang N, Liu T, Wang J, Xiao Y, Zhang Y, Dai J, Ma Z, Ma D. Si-Ni-San Reduces Hepatic Lipid Deposition in Rats with Metabolic Associated Fatty Liver Disease by AMPK/SIRT1 Pathway. Drug Des Devel Ther 2023; 17:3047-3060. [PMID: 37808345 PMCID: PMC10559901 DOI: 10.2147/dddt.s417378] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 09/21/2023] [Indexed: 10/10/2023] Open
Abstract
Background Metabolic associated fatty liver disease (MAFLD) is a chronic disease characterized by excessive lipid deposition in the liver without alcohol or other clear liver-damaging factors. AMP-activated protein kinase (AMPK)/silencing information regulator (SIRT)1 signaling pathway plays an important role in MAFLD development. Si-Ni-San (SNS), a traditional Chinese medicine, has shown reducing hepatic lipid deposition in MAFLD rats, however, the underlying mechanisms of SNS are barely understood. Purpose The aim of this research was to investigate the mechanisms of SNS in reducing hepatic lipid deposition in MAFLD rats by regulating AMPK/SIRT1 signaling pathways. Methods The components of SNS were determined by high performance liquid chromatography with mass spectrometry (HPLC-MS) analysis. MAFLD rats were induced by high-fat and high-cholesterol diet (HFHCD), and treated by SNS. SNS-containing serum and Compound C (AMPK inhibitor) were used to treat palmitic acid (PA)-induced HepG2 cells. To elucidate the potential mechanism, lipid synthesis-related proteins (SREBP-1c and FAS), fatty acid oxidation (PPARα and CPT-1), and AMPK/SIRT1 signaling pathway (p-AMPK and SIRT1) were assessed by Western blot. Results SNS improved serum lipid levels, liver function and reduced hepatic lipid deposition in MAFLD rats. SNS-containing serum reduced lipid deposition in PA-induced HepG2 cells. SNS could up-regulate protein expressions of PPARα, CPT-1, p-AMPK and SIRT1, and down-regulate protein expressions of SREBP-1c and FAS. Similar effects of SNS-containing serum were observed in PA-induced HepG2 cells. Meanwhile, Compound C weakened the therapeutic effects of SNS-containing serum on lipid deposition. Conclusion SNS could reduce hepatic lipid deposition by inhibiting lipid synthesis and promoting fatty acid oxidation, which might be related with activating the AMPK/SIRT1 signaling pathway. This study could provide a theoretical basis for the clinical use of SNS to treat MAFLD.
Collapse
Affiliation(s)
- Ning Zhang
- School of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, People’s Republic of China
| | - Tong Liu
- School of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, People’s Republic of China
| | - Jianan Wang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, People’s Republic of China
| | - Yingying Xiao
- School of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, People’s Republic of China
| | - Ying Zhang
- School of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, People’s Republic of China
| | - Jun Dai
- School of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, People’s Republic of China
| | - Zhihong Ma
- School of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, People’s Republic of China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Shijiazhuang, Hebei, 050200, People’s Republic of China
| | - Donglai Ma
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, People’s Republic of China
| |
Collapse
|
19
|
Choi KJ, Yoon MY, Kim JE, Yoon SS. Gut commensal Kineothrix alysoides mitigates liver dysfunction by restoring lipid metabolism and gut microbial balance. Sci Rep 2023; 13:14668. [PMID: 37674003 PMCID: PMC10482948 DOI: 10.1038/s41598-023-41160-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/22/2023] [Indexed: 09/08/2023] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), previously known as Non-Alcoholic Fatty Liver Disease, is a widespread liver condition characterized by excessive fat buildup in hepatocytes without significant alcohol consumption. Manipulation of the gut microbiome has been considered to prevent and improve the occurrence and progression of MASLD, particularly through the gut-liver axis. This study aimed to investigate the correlation between the gut microbiome and liver function and determine whether the gut microbiome can ameliorate MASLD. We comparatively analyzed the gut microbiome composition between mice fed normal chow and those fed a high-fat diet and observed that the abundance of Kineothrix alysoides decreased in the high-fat group. Further analysis showed that treatment with K. alysoides in the high-fat diet group led to decreased weight loss, and MASLD attenuation. Importantly, K. alysoides treatment attenuated MASLD in mice fed a high-fat, high-fructose diet (HFHF), which can cause advanced liver damage. Furthermore, administration of K. alysoides altered the gut microbial composition in the HFHF diet group and improved MASLD. Overall, these findings demonstrate the potential of K. alysoides in restoring gut health and facilitating lipid metabolism to prevent and treat MASLD.
Collapse
Affiliation(s)
- Kyoung Jin Choi
- Department of Microbiology and Immunology, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, South Korea
- Brain Korea 21 Project for Medical Sciences, Yonsei University College of Medicine, Seoul, South Korea
| | - Mi Young Yoon
- Department of Microbiology and Immunology, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, South Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Ji-Eun Kim
- Department of Microbiology and Immunology, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, South Korea.
- Institute of Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, South Korea.
| | - Sang Sun Yoon
- Department of Microbiology and Immunology, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, South Korea.
- Brain Korea 21 Project for Medical Sciences, Yonsei University College of Medicine, Seoul, South Korea.
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea.
- Institute of Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, South Korea.
- BioMe Inc., Seoul, South Korea.
| |
Collapse
|
20
|
Yang J, Zuo J, Deng Y, Zhang L, Yu H, Zhang C, Li Z, Li H. Antidiabetic activity of Tartary buckwheat protein-derived peptide AFYRW and its effects on protein glycosylation of pancreas in mice. Amino Acids 2023; 55:1063-1071. [PMID: 37341830 DOI: 10.1007/s00726-023-03294-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 06/11/2023] [Indexed: 06/22/2023]
Abstract
Diabetes Mellitus (DM) is one of the most important public health problems, and new antidiabetic drugs with fewer side effects are urgently needed. Here, we measured the antidiabetic effects of an antioxidant peptide (Ala-Phe-Tyr-Arg-Trp, AFYRW) from Tartary Buckwheat Albumin (TBA) in a high-fat diet/streptozotocin (HFD/STZ)-induced diabetic mouse model. The data showed that AFYRW suppressed hepatocyte steatosis and triglycerides while ameliorating insulin resistance in mice. Successively, the influence of AFYRW on aberrant protein glycosylation in diabetic mice was further investigated by lectin microarrays. The results suggested AFYRW could restore the expression of GalNAc, GalNAcα1-3Gal and GalNAcα1-3Galβ1-3/4Glc recognized by PTL-I, Siaα2-3Galβ1-4Glc(NAc)/Glc, Siaα2-3Gal, Siaα2-3 and Siaα2-3GalNAc recognized by MAL-II, terminating in GalNAcα/β1-3/6Gal recognized by WFA and αGalNAc, αGal, anti-A and B recognized by GSI-I to normal levels in the pancreas of HFD-STZ-induced diabetic mice. This work may provide new targets for the future discovery of potential biomarkers to evaluate the efficacy of food-derived antidiabetic drugs based on precise alterations of glycopatterns in DM.
Collapse
Affiliation(s)
- Jiajun Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Guizhou Medical University, Guiyang, 550004, People's Republic of China
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, 550004, Guizhou, People's Republic of China
| | - Jie Zuo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Yan Deng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Lilin Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Hanjie Yu
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, 710069, People's Republic of China
| | - Chen Zhang
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, 710069, People's Republic of China
| | - Zheng Li
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, 710069, People's Republic of China.
| | - Hongmei Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Guizhou Medical University, Guiyang, 550004, People's Republic of China.
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, 550004, Guizhou, People's Republic of China.
| |
Collapse
|
21
|
Shatta MA, El-Derany MO, Gibriel AA, El-Mesallamy HO. Rhamnetin ameliorates non-alcoholic steatosis and hepatocellular carcinoma in vitro. Mol Cell Biochem 2023; 478:1689-1704. [PMID: 36495373 PMCID: PMC10267014 DOI: 10.1007/s11010-022-04619-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 11/23/2022] [Indexed: 12/14/2022]
Abstract
Non-alcoholic fatty liver (NAFLD) is a widespread disease with various complications including Non-alcoholic steatohepatitis (NASH) that could lead to cirrhosis and ultimately hepatocellular carcinoma (HCC). Up till now there is no FDA approved drug for treatment of NAFLD. Flavonoids such as Rhamnetin (Rhm) have been ascribed effective anti-inflammatory and anti-oxidative properties. Thus, Rhm as a potent flavonoid could target multiple pathological cascades causing NAFLD to prevent its progression into HCC. NAFLD is a multifactorial disease and its pathophysiology is complex and is currently challenged by the 'Multiple-hit hypothesis' that includes wider range of comorbidities rather than previously established theory of 'Two-hit hypothesis'. Herein, we aimed at establishing reliable in vitro NASH models using different mixtures of variable ratios and concentrations of oleic acid (OA) and palmitic acid (PA) combinations using HepG2 cell lines. Moreover, we compared those models in the context of oil red staining, triglyceride levels and their altered downstream molecular signatures for genes involved in de novo lipogenesis, inflammation, oxidative stress and apoptotic machineries as well. Lastly, the effect of Rhm on NASH and HCC models was deeply investigated. Over the 10 NASH models tested, PA 500 µM concentration was the best model to mimic the molecular events of steatosis induced NAFLD. Rhm successfully ameliorated the dysregulated molecular events caused by the PA-induced NASH. Additionally, Rhm regulated inflammatory and oxidative machinery in the HepG2 cancerous cell lines. In conclusion, PA 500 µM concentration is considered an effective in vitro model to mimic NASH. Rhm could be used as a promising therapeutic modality against both NASH and HCC pathogenesis.
Collapse
Affiliation(s)
- Mahmoud A Shatta
- Department of Biochemistry, Faculty of Pharmacy, The British University in Egypt, Cairo, 11837, Egypt
| | - Marwa O El-Derany
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt.
| | - Abdullah A Gibriel
- Department of Biochemistry, Faculty of Pharmacy, The British University in Egypt, Cairo, 11837, Egypt
| | - Hala O El-Mesallamy
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
- Dean of Faculty of Pharmacy, Sinai University, North Sinai, 45518, Egypt
| |
Collapse
|
22
|
Alabdulaali B, Al-rashed F, Al-Onaizi M, Kandari A, Razafiarison J, Tonui D, Williams MR, Blériot C, Ahmad R, Alzaid F. Macrophages and the development and progression of non-alcoholic fatty liver disease. Front Immunol 2023; 14:1195699. [PMID: 37377968 PMCID: PMC10291618 DOI: 10.3389/fimmu.2023.1195699] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
The liver is the site of first pass metabolism, detoxifying and metabolizing blood arriving from the hepatic portal vein and hepatic artery. It is made up of multiple cell types, including macrophages. These are either bona fide tissue-resident Kupffer cells (KC) of embryonic origin, or differentiated from circulating monocytes. KCs are the primary immune cells populating the liver under steady state. Liver macrophages interact with hepatocytes, hepatic stellate cells, and liver sinusoidal endothelial cells to maintain homeostasis, however they are also key contributors to disease progression. Generally tolerogenic, they physiologically phagocytose foreign particles and debris from portal circulation and participate in red blood cell clearance. However as immune cells, they retain the capacity to raise an alarm to recruit other immune cells. Their aberrant function leads to the development of non-alcoholic fatty liver disease (NAFLD). NAFLD refers to a spectrum of conditions ranging from benign steatosis of the liver to steatohepatitis and cirrhosis. In NAFLD, the multiple hit hypothesis proposes that simultaneous influences from the gut and adipose tissue (AT) generate hepatic fat deposition and that inflammation plays a key role in disease progression. KCs initiate the inflammatory response as resident immune effectors, they signal to neighbouring cells and recruit monocytes that differentiated into recruited macrophages in situ. Recruited macrophages are central to amplifying the inflammatory response and causing progression of NAFLD to its fibro-inflammatory stages. Given their phagocytic capacity and their being instrumental in maintaining tissue homeostasis, KCs and recruited macrophages are fast-becoming target cell types for therapeutic intervention. We review the literature in the field on the roles of these cells in the development and progression of NAFLD, the characteristics of patients with NAFLD, animal models used in research, as well as the emerging questions. These include the gut-liver-brain axis, which when disrupted can contribute to decline in function, and a discussion on therapeutic strategies that act on the macrophage-inflammatory axis.
Collapse
Affiliation(s)
- Bader Alabdulaali
- Dasman Diabetes Institute, Kuwait City, Kuwait
- Ministry of Health, Kuwait City, Kuwait
| | | | - Mohammed Al-Onaizi
- Dasman Diabetes Institute, Kuwait City, Kuwait
- Department of Anatomy, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Anwar Kandari
- Dasman Diabetes Institute, Kuwait City, Kuwait
- Ministry of Health, Kuwait City, Kuwait
| | - Joanna Razafiarison
- INSERM UMR-S1151, CNRS UMR-S8253, Université Paris Cité, Institut Necker Enfants Malades, Paris, France
| | - Dorothy Tonui
- INSERM UMR-S1151, CNRS UMR-S8253, Université Paris Cité, Institut Necker Enfants Malades, Paris, France
| | | | - Camille Blériot
- INSERM UMR-S1151, CNRS UMR-S8253, Université Paris Cité, Institut Necker Enfants Malades, Paris, France
- Inserm U1015, Gustave Roussy, Villejuif, France
| | | | - Fawaz Alzaid
- Dasman Diabetes Institute, Kuwait City, Kuwait
- INSERM UMR-S1151, CNRS UMR-S8253, Université Paris Cité, Institut Necker Enfants Malades, Paris, France
| |
Collapse
|
23
|
Wang XX, Jin R, Li XH, Yang Q, Teng X, Liu FF, Wu N, Rao HY, Liu F. Collagen co-localized with macrovesicular steatosis better differentiates fibrosis progression in non-alcoholic fatty liver disease mouse models. Front Med (Lausanne) 2023; 10:1172058. [PMID: 37332758 PMCID: PMC10272541 DOI: 10.3389/fmed.2023.1172058] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 05/15/2023] [Indexed: 06/20/2023] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) is a global commonly occurring liver disease. However, its exact pathogenesis is not fully understood. The purpose of this study was to quantitatively evaluate the progression of steatosis and fibrosis by examining their distribution, morphology, and co-localization in NAFLD animal models. Methods Six mouse NAFLD groups were established: (1) western diet (WD) group; (2) WD with fructose in drinking water (WDF) group; (3) WDF + carbon tetrachloride (CCl4) group, WDF plus intraperitoneal injection of CCl4; (4) high-fat diet (HFD) group, (5) HFD with fructose (HFDF) group; and (6) HFDF + CCl4 group, HFDF plus intraperitoneal injection of CCl4. Liver tissue specimens from NAFLD model mice were collected at different time points. All the tissues were serially sectioned for histological staining and second-harmonic generation (SHG)/two-photon excitation fluorescence imaging (TPEF) imaging. The progression of steatosis and fibrosis was analyzed using SHG/TPEF quantitative parameters with respect to the non-alcoholic steatohepatitis Clinical Research Network scoring system. Results qSteatosis showed a good correlation with steatosis grade (R: 0.823-0.953, p < 0.05) and demonstrated high performance (area under the curve [AUC]: 0.617-1) in six mouse models. Based on their high correlation with histological scoring, qFibrosis containing four shared parameters (#LongStrPS, #ThinStrPS, #ThinStrPSAgg, and #LongStrPSDis) were selected to create a linear model that could accurately identify differences among fibrosis stages (AUC: 0.725-1). qFibrosis co-localized with macrosteatosis generally correlated better with histological scoring and had a higher AUC in six animal models (AUC: 0.846-1). Conclusion Quantitative assessment using SHG/TPEF technology can be used to monitor different types of steatosis and fibrosis progression in NAFLD models. The collagen co-localized with macrosteatosis could better differentiate fibrosis progression and might aid in developing a more reliable and translatable fibrosis evaluation tool for animal models of NAFLD.
Collapse
Affiliation(s)
- Xiao-Xiao Wang
- Peking University People’s Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, China
| | - Rui Jin
- Peking University People’s Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, China
| | - Xiao-He Li
- Peking University People’s Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, China
| | - Qiang Yang
- Hangzhou Choutu Technology Co., Ltd., Hangzhou, China
| | - Xiao Teng
- HistoIndex Pte Ltd, Singapore, Singapore
| | - Fang-Fang Liu
- Department of Pathology, Peking University People's Hospital, Beijing, China
| | - Nan Wu
- Peking University People’s Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, China
| | - Hui-Ying Rao
- Peking University People’s Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, China
| | - Feng Liu
- Peking University People’s Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, China
| |
Collapse
|
24
|
Helal KM, Cahyadi H, Taylor JN, Okajima A, Tabata K, Kumamoto Y, Mochizuki K, Itoh Y, Takamatsu T, Tanaka H, Fujita K, Komatsuzaki T, Harada Y. Raman imaging of rat nonalcoholic fatty liver tissues reveals distinct biomolecular states. FEBS Lett 2023. [PMID: 36807196 DOI: 10.1002/1873-3468.14600] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 12/24/2022] [Accepted: 01/08/2023] [Indexed: 02/20/2023]
Abstract
An essential challenge in diagnosing states of nonalcoholic fatty liver disease (NAFLD) is the early prediction of progression from nonalcoholic fatty liver (NAFL) to nonalcoholic steatohepatitis (NASH) before the disease progresses. Histological diagnoses of NAFLD rely on the appearance of anomalous tissue morphologies, and it is difficult to segment the biomolecular environment of the tissue through a conventional histopathological approach. Here, we show that hyperspectral Raman imaging provides diagnostic information on NAFLD in rats, as spectral changes among disease states can be detected before histological characteristics emerge. Our results demonstrate that Raman imaging of NAFLD can be a useful tool for histopathologists, offering biomolecular distinctions among tissue states that cannot be observed through standard histopathological means.
Collapse
Affiliation(s)
- Khalifa Mohammad Helal
- Graduate School of Life Science, Transdisciplinary Life Science Course, Hokkaido University, Sapporo, Japan.,Department of Mathematics, Comilla University, Cumilla, Bangladesh
| | - Harsono Cahyadi
- Department of Pathology and Cell Regulation, Kyoto Prefectural University of Medicine, Japan
| | - J Nicholas Taylor
- Research Institute for Electronic Science, Hokkaido University, Sapporo, Japan
| | - Akira Okajima
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Japan
| | - Koji Tabata
- Research Institute for Electronic Science, Hokkaido University, Sapporo, Japan
| | - Yasuaki Kumamoto
- Department of Pathology and Cell Regulation, Kyoto Prefectural University of Medicine, Japan
| | - Kentaro Mochizuki
- Department of Pathology and Cell Regulation, Kyoto Prefectural University of Medicine, Japan
| | - Yoshito Itoh
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Japan
| | - Tetsuro Takamatsu
- Department of Medical Photonics, Kyoto Prefectural University of Medicine, Japan
| | - Hideo Tanaka
- Department of Pathology and Cell Regulation, Kyoto Prefectural University of Medicine, Japan
| | - Katsumasa Fujita
- Department of Applied Physics, Osaka University, Japan.,Transdimensional Life Imaging Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Japan.,Advanced Photonics and Biosensing Open Innovation Laboratory, AIST-Osaka University, Japan
| | - Tamiki Komatsuzaki
- Graduate School of Life Science, Transdisciplinary Life Science Course, Hokkaido University, Sapporo, Japan.,Research Institute for Electronic Science, Hokkaido University, Sapporo, Japan.,Laboratoire Interdisciplinaire Carnot de Bourgogne, UMR 6303 CNRS-Univ. Bourgogne Franche-Comté, Dijon Cedex, France
| | - Yoshinori Harada
- Department of Pathology and Cell Regulation, Kyoto Prefectural University of Medicine, Japan
| |
Collapse
|
25
|
Chen Q, Lou Y. G protein-coupled receptor 39 alleviates mitochondrial dysfunction and hepatocyte lipid accumulation via SIRT1/Nrf2 signaling. J Bioenerg Biomembr 2023; 55:33-42. [PMID: 36525212 DOI: 10.1007/s10863-022-09953-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/03/2022] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Data in the GEO database (GSE63067) showed that G protein-coupled receptor 39 (GPR39) was down-regulated in tissues from patients with non-alcoholic fatty liver disease (NAFLD). It was intended to explore the mechanism of GPR39 in NAFLD. METHODS HepG2 cells were treated with a mixture of oleic acid and palmitic acid (OA/PA) to mimic NAFLD cell models. The level of GPR39 and the functions of GPR39 on cellular oxidative stress, lipid accumulation, the SIRT1/Nrf2 signaling and mitochondrial dysfunction were assessed. To verify the mediation of the SIRT1 signaling pathway in GPR39 regulation, cells were subjected to SIRT1 inhibitor EX-527 treatment. Afterwards, the abovementioned aspects of cells were all determined. RESULTS GPR39 presented a downward trend in response to OA/PA. GPR39 overexpression could suppress oxidative stress, lipid accumulation and activate the SIRT1/Nrf2 signaling. GPR39 overexpression likewise alleviated mitochondrial dysfunction, whereas EX-527 treatment disturbed the effects of GPR39 overexpression on these aspects. CONCLUSION The present study found that GPR39 reduced oxidative stress and maintained mitochondrial homeostasis in a cellular model of NAFLD, a process mediated by SIRT1/Nrf2 signaling.
Collapse
Affiliation(s)
- Qiang Chen
- Department of Gastroenterology, Sanmen County People's Hospital, 15 Taihe Road, Hairun Street, 317100, Taizhou, Zhejiang, China.
| | - Yifeng Lou
- Department of Infection, Sanmen County People's Hospital, 317100, Taizhou, Zhejiang, China
| |
Collapse
|
26
|
Shama S, Jang H, Wang X, Zhang Y, Shahin NN, Motawi TK, Kim S, Gawrieh S, Liu W. Phosphatidylethanolamines Are Associated with Nonalcoholic Fatty Liver Disease (NAFLD) in Obese Adults and Induce Liver Cell Metabolic Perturbations and Hepatic Stellate Cell Activation. Int J Mol Sci 2023; 24:ijms24021034. [PMID: 36674549 PMCID: PMC9861886 DOI: 10.3390/ijms24021034] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 12/28/2022] [Accepted: 01/03/2023] [Indexed: 01/07/2023] Open
Abstract
Pathogenesis roles of phospholipids (PLs) in nonalcoholic fatty liver disease (NAFLD) remain incompletely understood. This study investigated the role of PLs in the progression of NAFLD among obese individuals via studying the alterations in serum PL composition throughout the spectrum of disease progression and evaluating the effects of specific phosphatidylethanolamines (PEs) on FLD development in vitro. A total of 203 obese subjects, who were undergoing bariatric surgery, were included in this study. They were histologically classified into 80 controls (C) with normal liver histology, 93 patients with simple hepatic steatosis (SS), 16 with borderline nonalcoholic steatohepatitis (B-NASH) and 14 with progressive NASH (NASH). Serum PLs were profiled by automated electrospray ionization tandem mass spectrometry (ESI-MS/MS). HepG2 (hepatoma cells) and LX2 (immortalized hepatic stellate cells or HSCs) were used to explore the roles of PL in NAFLD/NASH development. Several PLs and their relative ratios were significantly associated with NAFLD progression, especially those involving PE. Incubation of HepG2 cells with two phosphatidylethanolamines (PEs), PE (34:1) and PE (36:2), resulted in significant inhibition of cell proliferation, reduction of mitochondrial mass and membrane potential, induction of lipid accumulation and mitochondrial ROS production. Meanwhile, treatment of LX2 cells with both PEs markedly increased cell activation and migration. These effects were associated with a significant change in the expression levels of genes involved in lipogenesis, lipid oxidation, autophagy, apoptosis, inflammation, and fibrosis. Thus, our study demonstrated that elevated level of PEs increases susceptibility to the disease progression of obesity associated NAFLD, likely through a causal cascade of impacts on the function of different liver cells.
Collapse
Affiliation(s)
- Samaa Shama
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
- Cell-Based Analysis Unit, Reference Laboratory, Egyptian Drug Authority, Cairo 12618, Egypt
| | - Hyejeong Jang
- Biostatistics and Bioinformatics Core, Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Xiaokun Wang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Yang Zhang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Nancy Nabil Shahin
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Tarek Kamal Motawi
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
- Correspondence: (T.K.M.); (W.L.); Tel.: +20-122-313-8667 (T.K.M.); +1-313-577-3375 (W.L.)
| | - Seongho Kim
- Biostatistics and Bioinformatics Core, Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Samer Gawrieh
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Wanqing Liu
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Correspondence: (T.K.M.); (W.L.); Tel.: +20-122-313-8667 (T.K.M.); +1-313-577-3375 (W.L.)
| |
Collapse
|
27
|
Jiang H, Mao T, Sun Z, Shi L, Han X, Zhang Y, Zhang X, Wang J, Hu J, Zhang L, Li J, Han H. Yinchen Linggui Zhugan decoction ameliorates high fat diet-induced nonalcoholic fatty liver disease by modulation of SIRT1/Nrf2 signaling pathway and gut microbiota. Front Microbiol 2022; 13:1001778. [PMID: 36578580 PMCID: PMC9791106 DOI: 10.3389/fmicb.2022.1001778] [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: 07/24/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022] Open
Abstract
Yinchen Linggui Zhugan decoction (YLZD) is an effective and classical traditional herbal prescription for treating the nonalcoholic fatty liver disease (NAFLD) and has been proven to be effective in the regulation of lipid metabolism disorder and attenuate inflammation for a NAFLD rat model. However, the exact underlying mechanism has not been elucidated. In the current study, a NAFLD rat model was established using a high-fat diet (HFD) for 10 weeks, followed by YLZD treatment with 1.92 g/kg/day for 4 weeks to explore the mechanisms of YLZD. Our results showed that YLZD decreased the hepatic lipid deposition, restored the liver tissue pathological lesions, inhibited the expression of oxidative stress, and decreased the inflammatory cytokines levels. Meanwhile, the genes and proteins expressions of SIRT1/Nrf2 signaling pathway together with downstream factors including HO-1 and NQO1 were elevated in the YLZD treated NAFLD rats. For further elaborating the upstream mechanism, short-chain fatty acids (SCFAs) in serum and feces were measured by liquid chromatograph mass spectrometer and gas chromatograph mass spectrometer, and the differences in gut microbiota of rats in each group were analyzed through high-throughput sequencing of 16S rRNA. The results demonstrated that the contents of butyric acid (BA) and total SCFAs in YLZD-treated NAFLD rats were significantly increased in serum and feces. 16S rRNA sequencing analysis illustrated that YLZD intervention led to a modification of the gut microbiota composition, with a decrease of Oribacterium, Lactobacillus and the ratio of Firmicutes/Bacteroides, as well as the increase in SCFAs-producing bacteria such as Christensenellaceae, Clostridia, Muribaculaceae, and Prevotellaceae. Spearman rank correlation analysis indicated that BA and total SCFAs were negatively co-related with oxidative stress-related factors and inflammatory cytokines, while they were positively co-related with SIRT1/Nrf2 pathway related genes and proteins. Furthermore, in vitro study confirmed that BA effectively reduced oxidative stress by activating SIRT1/Nrf2 signaling pathway in L02 cells. Together, the present data revealed YLZD could ameliorate HFD-induced NAFLD in rats by the modulation of SIRT1/Nrf2 signaling pathway and gut microbiota.
Collapse
Affiliation(s)
- Hui Jiang
- School of Graduate, Beijing University of Chinese Medicine, Beijing, China,Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Tangyou Mao
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Zhongmei Sun
- School of Graduate, Beijing University of Chinese Medicine, Beijing, China,Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Lei Shi
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiao Han
- School of Graduate, Beijing University of Chinese Medicine, Beijing, China,Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yang Zhang
- School of Graduate, Beijing University of Chinese Medicine, Beijing, China,Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaosi Zhang
- School of Graduate, Beijing University of Chinese Medicine, Beijing, China,Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jiali Wang
- School of Graduate, Beijing University of Chinese Medicine, Beijing, China,Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Juncong Hu
- School of Graduate, Beijing University of Chinese Medicine, Beijing, China,Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Liming Zhang
- School of Graduate, Beijing University of Chinese Medicine, Beijing, China,Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Junxiang Li
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China,*Correspondence: Junxiang Li, Haixiao Han
| | - Haixiao Han
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China,*Correspondence: Junxiang Li, Haixiao Han
| |
Collapse
|
28
|
Shedding light on non-alcoholic fatty liver disease: Pathogenesis, molecular mechanisms, models, and emerging therapeutics. Life Sci 2022; 312:121185. [PMID: 36375569 DOI: 10.1016/j.lfs.2022.121185] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 11/13/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disorder globally impacting an estimated 25% of the population associated with severe consequences such as cirrhosis, hepatocellular carcinoma (HCC), and overall mortality. Fatty liver disease is triggered through multiple pathways, but the most prominent cause is either diabetes or obesity, or a combination of both. Therefore, hepatic glucose, insulin and fatty acid signaling becomes a dire need to understand which is well elaborated in this review. This review summarizes the popular two-hit pathogenesis of NAFLD, the molecular mechanisms underlying hepatic insulin resistance. As fatty liver disease gets advanced, it requires in-vitro as well as in-vivo models closer to disease progression in humans for better understanding the pathological state and identifying a novel therapeutic target. This review summarizes in-vitro (2D cell-culture/co-culture, 3D spheroid/organoid/liver-on-a-chip) models as well as in-vivo (genetically/dietary/chemically induced fatty liver disease) research models. Fatty liver disease research has gathered lots of attention recently since there is no FDA approved therapy available so far. However, there have been numerous promising targets to treat fatty liver disease including potential therapeutic targets under clinical trials are listed in this review.
Collapse
|
29
|
Krüger M, Ruppelt A, Kappler B, Van Soest E, Samsom RA, Grinwis GCM, Geijsen N, Helms JB, Stijnen M, Kock LM, Rasponi M, Kooistra HS, Spee B. Normothermic Ex Vivo Liver Platform Using Porcine Slaughterhouse Livers for Disease Modeling. Bioengineering (Basel) 2022; 9:bioengineering9090471. [PMID: 36135018 PMCID: PMC9495507 DOI: 10.3390/bioengineering9090471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/25/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022] Open
Abstract
Metabolic and toxic liver disorders, such as fatty liver disease (steatosis) and drug-induced liver injury, are highly prevalent and potentially life-threatening. To allow for the study of these disorders from the early stages onward, without using experimental animals, we collected porcine livers in a slaughterhouse and perfused these livers normothermically. With our simplified protocol, the perfused slaughterhouse livers remained viable and functional over five hours of perfusion, as shown by hemodynamics, bile production, indocyanine green clearance, ammonia metabolism, gene expression and histology. As a proof-of-concept to study liver disorders, we show that an infusion of free fatty acids and acetaminophen results in early biochemical signs of liver damage, including reduced functionality. In conclusion, the present platform offers an accessible system to perform research in a functional, relevant large animal model while avoiding using experimental animals. With further improvements to the model, prolonged exposure could make this model a versatile tool for studying liver diseases and potential treatments.
Collapse
Affiliation(s)
- Melanie Krüger
- LifeTec Group BV, 5611 ZS Eindhoven, The Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CT Utrecht, The Netherlands
| | - Alicia Ruppelt
- LifeTec Group BV, 5611 ZS Eindhoven, The Netherlands
- Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano, 20133 Milan, Italy
- Correspondence:
| | | | | | - Roos Anne Samsom
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CT Utrecht, The Netherlands
| | - Guy C. M. Grinwis
- Veterinary Pathology Diagnostic Centre, Department of Biomedical Health Sciences, Faculty of Veterinary Medicine, Utrecht University, 3508 TD Utrecht, The Netherlands
| | - Niels Geijsen
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CT Utrecht, The Netherlands
| | - J. Bernd Helms
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, The Netherlands
| | - Marco Stijnen
- LifeTec Group BV, 5611 ZS Eindhoven, The Netherlands
| | - Linda M. Kock
- LifeTec Group BV, 5611 ZS Eindhoven, The Netherlands
- Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Marco Rasponi
- Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano, 20133 Milan, Italy
| | - Hans S. Kooistra
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CT Utrecht, The Netherlands
| | - Bart Spee
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CT Utrecht, The Netherlands
| |
Collapse
|
30
|
Akbari G, Mard SA, Savari F, Barati B, Sameri MJ. Characterization of diet based nonalcoholic fatty liver disease/nonalcoholic steatohepatitis in rodent models: Histological and biochemical outcomes. Histol Histopathol 2022; 37:813-824. [PMID: 35475465 DOI: 10.14670/hh-18-462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD), as the most common chronic liver disease, is rapidly increasing worldwide. This complex disorder can include simple liver steatosis to more serious stages of nonalcoholic fibrosis and steatohepatitis (NASH). One of the critical concerns in NASH research is selecting and confiding in relying on preclinical animal models and experimental methods that can accurately reflect the situation in human NASH. Recently, creating nutritional models of NASH with a closer dietary pattern in human has been providing reliable, simple, and reproducible tools that hope to create a better landscape for showing the recapitulation of disease pathophysiology. This review focuses on recent research on rodent models (mice, rats, and hamsters) in the induction of the dietary model of NAFLD /NASH. This research tries to compile the different dietary compositions of NASH, time frames required for disease development, and their impact on liver histological features as well as metabolic parameters.
Collapse
Affiliation(s)
- Ghaidafeh Akbari
- Medical Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Seyyed Ali Mard
- Clinical Sciences Research Institute, Alimentary Tract Research Center, Department of Physiology, The school of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Feryal Savari
- Department of Basic Sciences, Shoushtar Faculty of Medical Sciences, Shoushtar, Iran.
| | - Barat Barati
- Department of Radiologic Technology, Shoushtar Faculty of Medical Sciences, Shoushtar, Iran
| | - Maryam J Sameri
- Department of Physiology, The School of Medicine, Ahvaz Jundishpur University of Medical Sciences, Ahvaz, Iran
- Department of Physiology, The School of Medicine, Abadan University of Medical Sciences, Abadan, Iran
| |
Collapse
|
31
|
Lopez-Yus M, Lorente-Cebrian S, Del Moral-Bergos R, Hörndler C, Garcia-Sobreviela MP, Casamayor C, Sanz-Paris A, Bernal-Monterde V, Arbones-Mainar JM. Identification of novel targets in adipose tissue involved in non-alcoholic fatty liver disease progression. FASEB J 2022; 36:e22429. [PMID: 35792898 DOI: 10.1096/fj.202200118rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 06/06/2022] [Accepted: 06/10/2022] [Indexed: 12/14/2022]
Abstract
Obesity is a major risk factor for the development of Nonalcoholic fatty liver disease (NAFLD). We hypothesize that a dysfunctional subcutaneous white adipose tissue (scWAT) may lead to an accumulation of ectopic fat in the liver. Our aim was to investigate the molecular mechanisms involved in the causative role of scWAT in NALFD progression. We performed a RNA-sequencing analysis in a discovery cohort (n = 45) to identify genes in scWAT correlated with fatty liver index, a qualitative marker of liver steatosis. We then validated those targets in a second cohort (n = 47) of obese patients who had liver biopsies available. Finally, we obtained scWAT mesenchymal stem cells (MSCs) from 13 obese patients at different stages of NAFLD and established in vitro models of human MSC (hMSC)-derived adipocytes. We observed impaired adipogenesis in hMSC-derived adipocytes as liver steatosis increased, suggesting that an impaired adipogenic capacity is a critical event in the development of NAFLD. Four genes showed a differential expression pattern in both scWAT and hMSC-derived adipocytes, where their expression paralleled steatosis degree: SOCS3, DUSP1, SIK1, and GADD45B. We propose these genes as key players in NAFLD progression. They could eventually constitute potential new targets for future therapies against liver steatosis.
Collapse
Affiliation(s)
- Marta Lopez-Yus
- Adipocyte and Fat Biology Laboratory (AdipoFat), Translational Research Unit, University Hospital Miguel Servet, Instituto Aragones de Ciencias de la Salud (IACS), Zaragoza, Spain.,Instituto de Investigación Sanitaria (IIS) Aragon, Zaragoza, Spain
| | - Silvia Lorente-Cebrian
- Adipocyte and Fat Biology Laboratory (AdipoFat), Translational Research Unit, University Hospital Miguel Servet, Instituto Aragones de Ciencias de la Salud (IACS), Zaragoza, Spain.,Instituto de Investigación Sanitaria (IIS) Aragon, Zaragoza, Spain.,Departamento de Farmacología, Fisiología y Medicina Legal y Forense, Universidad de Zaragoza, Zaragoza, Spain.,Instituto Agroalimentario de Aragón (IA2) (Universidad de Zaragoza-CITA), Zaragoza, Spain
| | - Raquel Del Moral-Bergos
- Adipocyte and Fat Biology Laboratory (AdipoFat), Translational Research Unit, University Hospital Miguel Servet, Instituto Aragones de Ciencias de la Salud (IACS), Zaragoza, Spain.,Instituto de Investigación Sanitaria (IIS) Aragon, Zaragoza, Spain
| | - Carlos Hörndler
- Instituto de Investigación Sanitaria (IIS) Aragon, Zaragoza, Spain.,Pathology Department, Miguel Servet University Hospital, Zaragoza, Spain
| | - Maria Pilar Garcia-Sobreviela
- Adipocyte and Fat Biology Laboratory (AdipoFat), Translational Research Unit, University Hospital Miguel Servet, Instituto Aragones de Ciencias de la Salud (IACS), Zaragoza, Spain.,Instituto de Investigación Sanitaria (IIS) Aragon, Zaragoza, Spain
| | - Carmen Casamayor
- Instituto de Investigación Sanitaria (IIS) Aragon, Zaragoza, Spain.,Endocrine, Bariatric and Breast Surgery Unit, General and Digestive Surgery Department, Miguel Servet University Hospital, Zaragoza, Spain
| | - Alejandro Sanz-Paris
- Instituto de Investigación Sanitaria (IIS) Aragon, Zaragoza, Spain.,Nutrition Department, Miguel Servet University Hospital, Zaragoza, Spain
| | - Vanesa Bernal-Monterde
- Instituto de Investigación Sanitaria (IIS) Aragon, Zaragoza, Spain.,Gastroenterology Department, Miguel Servet University Hospital, Zaragoza, Spain
| | - Jose M Arbones-Mainar
- Adipocyte and Fat Biology Laboratory (AdipoFat), Translational Research Unit, University Hospital Miguel Servet, Instituto Aragones de Ciencias de la Salud (IACS), Zaragoza, Spain.,Instituto de Investigación Sanitaria (IIS) Aragon, Zaragoza, Spain.,CIBER Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto Salud Carlos III, Madrid, Spain
| |
Collapse
|
32
|
Zhang M, Zhang Y, Jiao X, Lai L, Qian Y, Sun B, Yang W. Identification and validation of immune related core transcription factors GTF2I in NAFLD. PeerJ 2022; 10:e13735. [PMID: 35891648 PMCID: PMC9308966 DOI: 10.7717/peerj.13735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 06/25/2022] [Indexed: 01/17/2023] Open
Abstract
Background Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease worldwide that endangers human health. Transcription factors (TFs) have gradually become hot spots for drug development in NAFLD for their impacts on metabolism. However, the specific TFs that regulate immune response in the development of NAFLD is not clear. This study aimed to investigate the TFs involved in the immune response of NAFLD and provide novel targets for drug development. Methods Microarray data were obtained from liver samples from 26 normal volunteers and 109 NAFLD patients using the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) were analyzed by limma package. Differentially expressed transcription factors (DETFs) were obtained on DEGs combined with Cistrome Cancer database. Immune signatures and pathways hallmark were identified by ssGSSEA and GSVA. The co-regulation network was constructed by the above results. Further, quantitative Real-time Polymerase Chain Reaction (qRT-PCR), Western blot (WB) and Immunohistochemistry (IHC) were used to validate the relationship between GTF2I and NAFLD. CIBERSORT analysis was performed to identify cell types to explore the relationship between differential expression of GTF2I and immune cell surface markers. Results A total of 617 DEGs and six DETFs (ESR1, CHD2, GTF2I, EGR1, HCFC1, SP2) were obtained by differential analysis. Immune signatures and pathway hallmarks were identified by ssGSSEA and GSVA. GTF2I and CHD2 were screened through the co-regulatory networks of DEGs, DETFs, immune signatures and pathway hallmarks. Furthermore, qRT-PCR, WB and IHC indicated that GTF2I but not CHD2 was significantly upregulated in NAFLD. Finally, in silico, our data confirmed that GTF2I has a wide impact on the immune profile by negatively regulating the expression of the chemokine receptor family (227/261, count of significance). Conclusion GTF2I plays a role in NAFLD by negatively regulating the chemokine receptor family, which affects the immune profile. This study may provide a potential target for the diagnosis or therapy of NAFLD.
Collapse
Affiliation(s)
- Minbo Zhang
- Department of Gastroenterology and Hepatology, Tongji Hospital of Tongji University, Shanghai, China
| | - Yu Zhang
- Department of Gastroenterology and Hepatology, Tongji Hospital of Tongji University, Shanghai, China
| | - Xiaoxiao Jiao
- Department of Gastroenterology and Hepatology, Tongji Hospital of Tongji University, Shanghai, China
| | - Linying Lai
- Department of Gastroenterology and Hepatology, Tongji Hospital of Tongji University, Shanghai, China
| | - Yiting Qian
- Department of Gastroenterology and Hepatology, Tongji Hospital of Tongji University, Shanghai, China
| | - Bo Sun
- Department of Gastroenterology and Hepatology, Tongji Hospital of Tongji University, Shanghai, China
| | - Wenzhuo Yang
- Department of Gastroenterology and Hepatology, Tongji Hospital of Tongji University, Shanghai, China
| |
Collapse
|
33
|
Su Y, Kang Y, Yi J, Lin Q, Zhang C, Lin Z, Yan Z, Qu J, Liu J. Isoschaftoside Reverses Nonalcoholic Fatty Liver Disease via Activating Autophagy In Vivo and In Vitro. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:2122563. [PMID: 35795282 PMCID: PMC9252632 DOI: 10.1155/2022/2122563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/31/2022] [Accepted: 06/01/2022] [Indexed: 11/18/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common metabolic liver disease globally, and the incidence of NAFLD has been increasing rapidly year by year. Currently, there is no effective pharmacotherapy for NAFLD. Therefore, studies are urgently needed to explore therapeutic drugs for NAFLD. In this study, we show that isoschaftoside (ISO) dramatically reduces lipid deposition in cells. Meanwhile, ISO treatment reverses the NAFLD and reduces hepatic steatosis in mice. Importantly, we reveal that ISO suppresses the expression of light-chain 3-II (LC3-II) and SQSTM1/p62 in palmitic acid (PA) induced autophagy inhibition in the cell model and the NAFLD mouse model, which suggests that ISO might reverse NAFLD through regulating autophagy flux. We propose that ISO might alleviate hepatic steatosis in NAFLD via regulating autophagy machinery. Consequently, our study suggests that ISO might be of potential clinical value in the field of NAFLD therapy. ISO might have the potential for future therapeutic application.
Collapse
Affiliation(s)
- Yanze Su
- Department of Clinical Medicine, Weifang Medical University, Weifang 261031, China
- Department of Hepatobiliary and Pancreatic Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Yixing Kang
- Department of Clinical Medicine, Weifang Medical University, Weifang 261031, China
- Department of Hepatobiliary and Pancreatic Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Jing Yi
- Department of Hepatobiliary and Pancreatic Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Qirui Lin
- Department of Hepatobiliary and Pancreatic Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Chaochuang Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Zewei Lin
- Department of Hepatobiliary and Pancreatic Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Zilong Yan
- Department of Hepatobiliary and Pancreatic Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Jianhua Qu
- Department of Hepatobiliary and Pancreatic Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Jikui Liu
- Department of Hepatobiliary and Pancreatic Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| |
Collapse
|
34
|
Ajah AA, Lembede BW, Nkomozepi P, Erlwanger KH, Nyakudya TT. Neonatal Oral Administration of Chrysin Prevents Long-Term Development of Non-Alcoholic Fatty Liver Disease in a Sexually Dimorphic Manner in Fructose Nurtured Sprague Dawley Rats. Life (Basel) 2022; 12:life12060790. [PMID: 35743821 PMCID: PMC9225280 DOI: 10.3390/life12060790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/30/2022] [Accepted: 05/23/2022] [Indexed: 12/03/2022] Open
Abstract
High-fructose diets are linked with the development of non-alcoholic fatty liver disease (NAFLD), the management of which is a burden to society. Interventions with phytochemicals in the early postnatal period may prevent fructose-induced NAFLD later in adulthood. We investigated the protective potential of chrysin against fructose-induced NAFLD. Four-day-old male and female suckling Sprague Dawley rats (N = 112) were randomly grouped and orally gavaged daily with distilled water (negative Control-Cn + W), chrysin(Chr-100 mg/kg), fructose-solution (Fr-20% w/v), and Chr + Fr between postnatal day (PND) 4 and 21 and then weaned onto normal rat chow and plain drinking water to PND 55. From PND 56 to 130, half of the rats continued on plain water, and the rest had Fr as drinking fluid. Terminally, the liver tissue was collected, and the lipid content was determined and histologically assessed for NAFLD. Dietary Fr induced an increased hepatic lipid content (p = 0.0001 vs. Cn + W) both sexes, and it was only attenuated by neonatal Chr in female rats (p < 0.05). Histologically, there was increased microvesicular steatosis (p = 0.0001 vs. Cn + W) in both sexes, and it was prevented by neonatal Chr (p > 0.05). Fr caused macrovesicular steatosis (p = 0.01 vs. Cn + W) in females only, and chrysin did not prevent it (p > 0.05). Fr induced hepatocellular hypertrophy, and inflammation was observed in females only (p = 0.01 vs. Cn + W), and this was prevented by Chr (p > 0.05). The collagen area fraction was increased by Fr (p = 0.02 (males) and p = 0.04 (females) vs. Cn + W, respectively; however, chrysin did not prevent this (p > 0.05). Neonatal chrysin prevented some of the deleterious effects of the high-fructose diet on the liver, suggesting that chrysin should be further explored as a strategic prophylactic neonatal intervention against high-fructose-diet-induced NAFLD.
Collapse
Affiliation(s)
- Austin A. Ajah
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa; (B.W.L.); (K.H.E.)
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, University of Port Harcourt, P.M.B. 5323, Choba, Port Harcourt 500102, Nigeria
- Correspondence:
| | - Busisani W. Lembede
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa; (B.W.L.); (K.H.E.)
| | - Pilani Nkomozepi
- Department of Human Anatomy and Physiology, Faculty of Health Sciences, University of Johannesburg, Corner Beit and Siemert Street, Doornfontein, Johannesburg 2094, South Africa; (P.N.); (T.T.N.)
| | - Kennedy H. Erlwanger
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa; (B.W.L.); (K.H.E.)
| | - Trevor T. Nyakudya
- Department of Human Anatomy and Physiology, Faculty of Health Sciences, University of Johannesburg, Corner Beit and Siemert Street, Doornfontein, Johannesburg 2094, South Africa; (P.N.); (T.T.N.)
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Private Bag X323, Gezina, Pretoria 0031, South Africa
| |
Collapse
|
35
|
Safran M, Masoud R, Sultan M, Tachlytski I, Chai Gadot C, Pery R, Balint-Lahat N, Pappo O, Buzaglo N, Ben-Ari Z. Extracellular Vesicular Transmission of miR-423-5p from HepG2 Cells Inhibits the Differentiation of Hepatic Stellate Cells. Cells 2022; 11:cells11101715. [PMID: 35626751 PMCID: PMC9139792 DOI: 10.3390/cells11101715] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 02/04/2023] Open
Abstract
Liver fibrosis (LF) is a major cause of morbidity and mortality worldwide. Hepatic stellate cells (HSCs) are the primary source of extracellular matrix in the liver and their activation is a central event in LF development. Extracellular vesicles (EVs) are intercellular communication agents, which play important roles in physiological processes in chronic liver diseases. The aim of this study was to examine the crosstalk between hepatocytes and HSCs mediated by hepatocyte-secreted EVs. EVs were purified from primary mouse hepatocytes, HepG2 cell lines, under normal or stressed conditions. The effect of EVs on primary HSCs (pHSCs) differentiation was evaluated by measuring of differentiation markers. In addition, their impact on the carbon tetrachloride (CCl4)-induced fibrosis mouse model was evaluated. The results demonstrated that HepG2-EVs regulate HSC differentiation and that under stress conditions, promoted pHSCs differentiation into the myofibroblast phenotype. The evaluation of miRNA sequences in the HepG2 secreted EVs demonstrated high levels of miR-423-5p. The examination of EV cargo following stress conditions identified a significant reduction of miR-423-5p in HepG2-EVs relative to HepG2-EVs under normal conditions. In addition, pHSCs transfected with miR-423-5p mimic and exhibit lower mRNA levels of alpha smooth muscle actin and Collagen type 1 alpha, and the mRNA expression level of genes targeted the family with sequence-similarity-3 (FAM3) and Monoacylglycerol lipase (Mgll). This study strengthened the hypothesis that EVs are involved in LF and that their cargo changes in stress conditions. In addition, miR-423-5p was shown to be involved in HSCs differentiation and hence, fibrosis development.
Collapse
Affiliation(s)
- Michal Safran
- Liver Diseases Center, Chaim Sheba Medical Center Tel-Hashomer, Ramat-Gan 5262000, Israel; (M.S.); (R.M.); (M.S.); (I.T.); (C.C.G.); (N.B.)
| | - Rula Masoud
- Liver Diseases Center, Chaim Sheba Medical Center Tel-Hashomer, Ramat-Gan 5262000, Israel; (M.S.); (R.M.); (M.S.); (I.T.); (C.C.G.); (N.B.)
| | - Maya Sultan
- Liver Diseases Center, Chaim Sheba Medical Center Tel-Hashomer, Ramat-Gan 5262000, Israel; (M.S.); (R.M.); (M.S.); (I.T.); (C.C.G.); (N.B.)
| | - Irena Tachlytski
- Liver Diseases Center, Chaim Sheba Medical Center Tel-Hashomer, Ramat-Gan 5262000, Israel; (M.S.); (R.M.); (M.S.); (I.T.); (C.C.G.); (N.B.)
| | - Chofit Chai Gadot
- Liver Diseases Center, Chaim Sheba Medical Center Tel-Hashomer, Ramat-Gan 5262000, Israel; (M.S.); (R.M.); (M.S.); (I.T.); (C.C.G.); (N.B.)
| | - Ron Pery
- Department of General Surgery, Chaim Sheba Medical Center Tel-Hashomer, Ramat-Gan 5262000, Israel;
| | - Nora Balint-Lahat
- Pathology Department, Chaim Sheba Medical Center Tel-Hashomer, Ramat-Gan 5262000, Israel; (N.B.-L.); (O.P.)
| | - Orit Pappo
- Pathology Department, Chaim Sheba Medical Center Tel-Hashomer, Ramat-Gan 5262000, Israel; (N.B.-L.); (O.P.)
| | - Nahum Buzaglo
- Liver Diseases Center, Chaim Sheba Medical Center Tel-Hashomer, Ramat-Gan 5262000, Israel; (M.S.); (R.M.); (M.S.); (I.T.); (C.C.G.); (N.B.)
| | - Ziv Ben-Ari
- Liver Diseases Center, Chaim Sheba Medical Center Tel-Hashomer, Ramat-Gan 5262000, Israel; (M.S.); (R.M.); (M.S.); (I.T.); (C.C.G.); (N.B.)
- Pathology Sackler School of Medicine, Tel Aviv University, Tel-Aviv 6329302, Israel
- Correspondence: ; Tel.: +972-3-5307180
| |
Collapse
|
36
|
Rajapaksha IG, Gunarathne LS, Asadi K, Laybutt R, Andrikopoulous S, Alexander IE, Watt MJ, Angus PW, Herath CB. Angiotensin Converting Enzyme-2 Therapy Improves Liver Fibrosis and Glycemic Control in Diabetic Mice With Fatty Liver. Hepatol Commun 2022; 6:1056-1072. [PMID: 34951153 PMCID: PMC9035567 DOI: 10.1002/hep4.1884] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 11/29/2021] [Accepted: 12/04/2021] [Indexed: 12/26/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver disease and is frequently associated with type 2 diabetes. However, there is no specific medical therapy to treat this condition. Angiotensin-converting enzyme 2 (ACE2) of the protective renin angiotensin system generates the antifibrotic peptide angiotensin-(1-7) from profibrotic angiotensin II peptide. In this study, we investigated the therapeutic potential of ACE2 in diabetic NAFLD mice fed a high-fat (20%), high-cholesterol (2%) diet for 40 weeks. Mice were given a single intraperitoneal injection of ACE2 using an adeno-associated viral vector at 30 weeks of high-fat, high-cholesterol diet (15 weeks after induction of diabetes) and sacrificed 10 weeks later. ACE2 significantly reduced liver injury and fibrosis in diabetic NAFLD mice compared with the control vector injected mice. This was accompanied by reductions in proinflammatory cytokine expressions, hepatic stellate cell activation, and collagen 1 expression. Moreover, ACE2 therapy significantly increased islet numbers, leading to an increased insulin protein content in β-cells and plasma insulin levels with subsequent reduction in plasma glucose levels compared with controls. Conclusion: We conclude that ACE2 gene therapy reduces liver fibrosis and hyperglycemia in diabetic NAFLD mice and has potential as a therapy for patients with NAFLD with diabetes.
Collapse
Affiliation(s)
- Indu G Rajapaksha
- Department of MedicineThe University of MelbourneAustin HealthHeidelbergVICAustralia
| | - Lakmie S Gunarathne
- Department of MedicineThe University of MelbourneAustin HealthHeidelbergVICAustralia
| | | | - Ross Laybutt
- Garvan Institute of Medical ResearchSydneyNSWAustralia.,St. Vincent's Clinical SchoolUniversity of New South WalesSydneyNSWAustralia
| | - Sof Andrikopoulous
- Department of MedicineThe University of MelbourneAustin HealthHeidelbergVICAustralia
| | - Ian E Alexander
- School of MedicineUniversity of SydneyChildren's Medical Research InstituteSydneyNSWAustralia
| | - Mathew J Watt
- Department Anatomy and PhysiologyThe University of MelbourneMelbourneVICAustralia
| | - Peter W Angus
- Department of MedicineThe University of MelbourneAustin HealthHeidelbergVICAustralia.,Department GastroenterologyAustin HealthHeidelbergVICAustralia
| | - Chandana B Herath
- Department of MedicineThe University of MelbourneAustin HealthHeidelbergVICAustralia.,South Western Sydney Clinical SchoolFaculty of MedicineUniversity of New South WalesSydneyNSWAustralia.,Ingham Institute for Applied Medical ResearchLiverpoolNSWAustralia
| |
Collapse
|
37
|
Kassotis CD, Vom Saal FS, Babin PJ, Lagadic-Gossmann D, Le Mentec H, Blumberg B, Mohajer N, Legrand A, Munic Kos V, Martin-Chouly C, Podechard N, Langouët S, Touma C, Barouki R, Kim MJ, Audouze K, Choudhury M, Shree N, Bansal A, Howard S, Heindel JJ. Obesity III: Obesogen assays: Limitations, strengths, and new directions. Biochem Pharmacol 2022; 199:115014. [PMID: 35393121 PMCID: PMC9050906 DOI: 10.1016/j.bcp.2022.115014] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 12/11/2022]
Abstract
There is increasing evidence of a role for environmental contaminants in disrupting metabolic health in both humans and animals. Despite a growing need for well-understood models for evaluating adipogenic and potential obesogenic contaminants, there has been a reliance on decades-old in vitro models that have not been appropriately managed by cell line providers. There has been a quick rise in available in vitro models in the last ten years, including commercial availability of human mesenchymal stem cell and preadipocyte models; these models require more comprehensive validation but demonstrate real promise in improved translation to human metabolic health. There is also progress in developing three-dimensional and co-culture techniques that allow for the interrogation of a more physiologically relevant state. While diverse rodent models exist for evaluating putative obesogenic and/or adipogenic chemicals in a physiologically relevant context, increasing capabilities have been identified for alternative model organisms such as Drosophila, C. elegans, zebrafish, and medaka in metabolic health testing. These models have several appreciable advantages, including most notably their size, rapid development, large brood sizes, and ease of high-resolution lipid accumulation imaging throughout the organisms. They are anticipated to expand the capabilities of metabolic health research, particularly when coupled with emerging obesogen evaluation techniques as described herein.
Collapse
Affiliation(s)
- Christopher D Kassotis
- Institute of Environmental Health Sciences and Department of Pharmacology, Wayne State University, Detroit, MI 48202, United States.
| | - Frederick S Vom Saal
- Division of Biological Sciences, The University of Missouri, Columbia, MO 65211, United States
| | - Patrick J Babin
- Department of Life and Health Sciences, University of Bordeaux, INSERM, Pessac, France
| | - Dominique Lagadic-Gossmann
- Univ Rennes, Inserm, EHESP, Irset (Research Institute for Environmental and Occupational Health) - UMR_S 1085, 35 000 Rennes, France
| | - Helene Le Mentec
- Univ Rennes, Inserm, EHESP, Irset (Research Institute for Environmental and Occupational Health) - UMR_S 1085, 35 000 Rennes, France
| | - Bruce Blumberg
- Department of Developmental and Cell Biology, The University of California, Irvine, Irvine CA 92697, United States
| | - Nicole Mohajer
- Department of Developmental and Cell Biology, The University of California, Irvine, Irvine CA 92697, United States
| | - Antoine Legrand
- Univ Rennes, Inserm, EHESP, Irset (Research Institute for Environmental and Occupational Health) - UMR_S 1085, 35 000 Rennes, France
| | - Vesna Munic Kos
- Department of Physiology and Pharmacology, Karolinska Institute, Solna, Sweden
| | - Corinne Martin-Chouly
- Univ Rennes, Inserm, EHESP, Irset (Research Institute for Environmental and Occupational Health) - UMR_S 1085, 35 000 Rennes, France
| | - Normand Podechard
- Univ Rennes, Inserm, EHESP, Irset (Research Institute for Environmental and Occupational Health) - UMR_S 1085, 35 000 Rennes, France
| | - Sophie Langouët
- Univ Rennes, Inserm, EHESP, Irset (Research Institute for Environmental and Occupational Health) - UMR_S 1085, 35 000 Rennes, France
| | - Charbel Touma
- Univ Rennes, Inserm, EHESP, Irset (Research Institute for Environmental and Occupational Health) - UMR_S 1085, 35 000 Rennes, France
| | - Robert Barouki
- Department of Biochemistry, University of Paris, INSERM, Paris, France
| | - Min Ji Kim
- Sorbonne Paris Nord University, Bobigny, INSERM U1124 (T3S), Paris, France
| | | | - Mahua Choudhury
- Department of Pharmaceutical Sciences, Texas A & M University, College Station, TX 77843, United States
| | - Nitya Shree
- Department of Pharmaceutical Sciences, Texas A & M University, College Station, TX 77843, United States
| | - Amita Bansal
- College of Health & Medicine, Australian National University, Canberra, ACT, 2611, Australia
| | - Sarah Howard
- Healthy Environment and Endocrine Disruptor Strategies, Commonweal, Bolinas, CA 92924, United States
| | - Jerrold J Heindel
- Healthy Environment and Endocrine Disruptor Strategies, Commonweal, Bolinas, CA 92924, United States
| |
Collapse
|
38
|
In vitro ballooned hepatocytes can be produced by primary human hepatocytes and hepatic stellate cell sheets. Sci Rep 2022; 12:5341. [PMID: 35351975 PMCID: PMC8964766 DOI: 10.1038/s41598-022-09428-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 03/17/2022] [Indexed: 11/17/2022] Open
Abstract
Despite the increasing prevalence of Nonalcoholic steatohepatitis (NASH) worldwide, there is no effective treatment available for this disease. “Ballooned hepatocyte” is a characteristic finding in NASH and is correlated with disease prognosis, but their mechanisms of action are poorly understood; furthermore, neither animal nor in vitro models of NASH have been able to adequately represent ballooned hepatocytes. Herein, we engineered cell sheets to develop a new in vitro model of ballooned hepatocytes. Primary human hepatocytes (PHH) and Hepatic stellate cells (HSC) were co-cultured to produce cell sheets, which were cultured in glucose and lipid containing medium, following which histological and functional analyses were performed. Histological findings showed hepatocyte ballooning, accumulation of fat droplets, abnormal cytokeratin arrangement, and the presence of Mallory–Denk bodies and abnormal organelles. These findings are similar to those of ballooned hepatocytes in human NASH. Functional analysis showed elevated levels of TGFβ-1, SHH, and p62, but not TNF-α, IL-8. Exposure of PHH/HSC sheets to a glucolipotoxicity environment induces ballooned hepatocyte without inflammation. Moreover, fibrosis is an important mechanism underlying ballooned hepatocytes and could be the basis for the development of a new in vitro NASH model with ballooned hepatocytes.
Collapse
|
39
|
Cominguez DC, Park YJ, Kang YM, Nugroho A, Kim S, An HJ. Clitorin ameliorates western diet-induced hepatic steatosis by regulating lipogenesis and fatty acid oxidation in vivo and in vitro. Sci Rep 2022; 12:4154. [PMID: 35264693 PMCID: PMC8907210 DOI: 10.1038/s41598-022-07937-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 02/28/2022] [Indexed: 12/02/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is usually correlated with metabolic diseases, such as obesity, insulin resistance, and hyperglycemia. Herein, we investigated the inhibitory effects and underlying governing mechanism of clitorin in a western diet (WD)-induced hepatic steatosis mouse model, and in oleic acid-stimulated HepG2 cells. Male C57BL/6 mice were fed a normal diet, WD, WD + 10 or 20 mg/kg orlistat, and WD + 10 or 20 mg/kg clitorin. HepG2 cells were treated with 1 mM oleic acid to induce lipid accumulation with or without clitorin. Clitorin significantly alleviated body weight gain and hepatic steatosis features (NAFLD activity score, micro-, and macro-vesicular steatosis) in WD-induced hepatic steatosis mice. Additionally, clitorin significantly decreased protein expressions of sterol regulatory element-binding protein 1 (SREBP1), peroxisome proliferator-activated receptor γ (PPARγ), and CCAAT/enhancer binding protein α (C/EBPα) in WD-induced hepatic steatosis mice. Moreover, clitorin significantly diminished the mRNA levels of SREBP1, acetyl-CoA carboxylase (ACC), fatty acid synthase (FAS), and hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR) and enhanced the mRNA levels of peroxisome proliferator-activated receptor α (PPARα) and carnitine palmitoyltranserase-1 (CTP-1), as well as adenosine monophosphate-activated protein kinase (AMPK) in the liver of WD-induced hepatic steatosis mice and oleic acid-stimulated HepG2 cells. Overall, our findings demonstrated that clitorin can be a potentially efficacious candidate for NAFLD management.
Collapse
Affiliation(s)
- Divina C Cominguez
- Department of Pharmacology, College of Korean Medicine, Sangji University, Wonju, Gangwon-do, 26339, Republic of Korea
| | - Yea-Jin Park
- Department of Pharmacology, College of Korean Medicine, Sangji University, Wonju, Gangwon-do, 26339, Republic of Korea
| | - Yun-Mi Kang
- Department of Pharmacology, College of Korean Medicine, Sangji University, Wonju, Gangwon-do, 26339, Republic of Korea
| | - Agung Nugroho
- Department of Agro-Industrial Technology, Lambung Mangkurat University, Banjarbaru, Indonesia
| | - Suhyun Kim
- Department of Obstetrics & Gynecology College of Korean Medicine, Sangji University, Wonju-si, Gangwon-do, 26339, Republic of Korea
| | - Hyo-Jin An
- Department of Pharmacology, College of Korean Medicine, Sangji University, Wonju, Gangwon-do, 26339, Republic of Korea.
| |
Collapse
|
40
|
Kim JH, Kim KM, Yang JH, Cho SS, Lee JH, Ki SH. Regulated in Development and DNA Damage Response 1 Protects Hepatocytes Against Palmitate-induced Lipotoxicity. BIOTECHNOL BIOPROC E 2022. [DOI: 10.1007/s12257-021-0140-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
41
|
Wang J, Du MM, Du Y, Li JX. HA-20 prevents hepatocyte steatosis in metabolic-associated fatty liver disease via regulating Ca 2+ relative signalling pathways. Eur J Pharmacol 2022; 921:174838. [PMID: 35218717 DOI: 10.1016/j.ejphar.2022.174838] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 02/09/2022] [Accepted: 02/15/2022] [Indexed: 11/03/2022]
Abstract
Metabolic-associated fatty liver disease (MAFLD) is caused by hepatocyte steatosis and is associated with obesity, type II diabetes, and heart disease. There are currently no effective drugs to treat MAFLD. This study explored the effect of HA-20, an oleanolic acid derivative, on hepatocyte steatosis in MAFLD. HepG2, L02, and AML12 cells were developed using oleic acid for in vitro MAFLD cell assays, and a high-fat diet + high-fructose diet-induced (HFHF) MAFLD mouse model was established for in vivo studies. The results demonstrated that HA-20 prevented hepatocyte steatosis in cell assays and caused 26.3, 57.7 and 70.0% inhibition of triglyceride (TG) levels in the 5.0, 10.0 and 20.0 μM HA-20 groups, respectively. The EC50 values of HA-20 treatment in HepG2, L02 and AML12 cells were 9.7 ± 0.6 μM, 42.4 ± 3.5 μM and 71.0 ± 14.7 μM, respectively. HA-20 also prevented hepatocyte steatosis in the MAFLD mouse model, the liver triglyceride contents were 2.3 ± 0.4 and 1.5 ± 0.2 mmol/L in the 2.5 and 5.0 mg/kg/day HA-20 groups, lower than 6.2 ± 0.7 mmol/L in the HFHF group and 3.3 ± 0.4 mmol/L in the metformin group. Further mechanistic investigation revealed that HA-20 increased the phosphorylation of calmodulin-dependent protein kinase kinase (p-CaMKK) and the phosphorylation of AMP-activated protein kinase (p-AMPK), at least partially by increasing intracellular Ca2+ concentration, which suppressed lipogenesis and enhanced β-oxidation. Our findings provide new insight into preventing MAFLD by increasing Ca2+ and suggest that HA-20 possesses therapeutic potential for MAFLD management.
Collapse
Affiliation(s)
- Jie Wang
- State Key Laboratory of Analytical Chemistry for Life Science, Collaborative Innovation Centre of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, PR China
| | - Ming-Ming Du
- State Key Laboratory of Analytical Chemistry for Life Science, Collaborative Innovation Centre of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, PR China
| | - Yun Du
- State Key Laboratory of Analytical Chemistry for Life Science, Collaborative Innovation Centre of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, PR China
| | - Jian-Xin Li
- State Key Laboratory of Analytical Chemistry for Life Science, Collaborative Innovation Centre of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, PR China.
| |
Collapse
|
42
|
Yang X, Deng Y, Tu Y, Feng D, Liao W. Nobiletin mitigates NAFLD via lipophagy and inflammation. Food Funct 2022; 13:10186-10199. [DOI: 10.1039/d2fo01682f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD), an increasingly serious health issue in the world, was characterized as lipid metabolic disorder without a satisfactory treatment. Nobiletin (NOB), a citrus flavonoid, was considered...
Collapse
|
43
|
Yang D, Jeong H, Hwang SM, Kim JW, Moon HW, Lee YE, Oh HB, Park CB, Kim B. Oral administration of Jinan Red Ginseng and licorice extract mixtures ameliorates nonalcoholic steatohepatitis by modulating lipogenesis. J Ginseng Res 2022; 46:126-137. [PMID: 35058729 PMCID: PMC8753527 DOI: 10.1016/j.jgr.2021.05.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 12/20/2022] Open
Abstract
Background Nonalcoholic steatohepatitis (NASH) is one of the main chronic liver diseases. NASH is identified by lipid accumulation, inflammation, and fibrosis. Jinan Red Ginseng (JRG) and licorice have been widely used because of their anti-inflammatory and hepatoprotective effects. Hence, this study assessed JRG and licorice extract mixtures' effects on NASH progression. Methods Palmitic acid (PA) and the western diet (WD) plus, high glucose-fructose water were used to induce in vitro and in vivo NASH. Mice were orally administered with JRG-single (JRG-S) and JRG-mixtures (JRG-M; JRG-S + licorice) at 0, 50, 100, 200 or 400 mg/kg/day once a day during the last half-period of diet feeding. Results JRG-S and JRG-M reduced NASH-related pathologies in WD-fed mice. JRG-S and JRG-M consistently decreased the mRNA level of genes related with inflammation, fibrosis, and lipid metabolism. The treatment of JRG-S and JRG-M also diminished the SREBP-1c protein levels and the p-AMPK/AMPK ratio. The FAS protein levels were decreased by JRG-M treatment both in vivo and in vitro but not JRG-S. Conclusion JRG-M effectively reduced lipogenesis by modulating AMPK downstream signaling. Our findings suggest that this mixture can be used as a prophylactic or therapeutic alternative for the remedy of NASH.
Collapse
Affiliation(s)
- Daram Yang
- Biosafety Research Institute and Laboratory of Pathology, College of Veterinary Medicine, Jeonbuk National University, Iksan-si, Jeollabuk-do, Republic of Korea
| | - Hyuneui Jeong
- Biosafety Research Institute and Laboratory of Pathology, College of Veterinary Medicine, Jeonbuk National University, Iksan-si, Jeollabuk-do, Republic of Korea
| | - Seung-Mi Hwang
- Department of Efficacy Study, Institute of Jinan Red Ginseng, Jinan-gun, Jeollabuk-do, Republic of Korea
- Department of Food Science and Technology, Jeonbuk National University, Deokjin-gu, Jeonju-si, Jeollabuk-do, Republic of Korea
| | - Jong-Won Kim
- Biosafety Research Institute and Laboratory of Pathology, College of Veterinary Medicine, Jeonbuk National University, Iksan-si, Jeollabuk-do, Republic of Korea
| | - Hee-Won Moon
- Biosafety Research Institute and Laboratory of Pathology, College of Veterinary Medicine, Jeonbuk National University, Iksan-si, Jeollabuk-do, Republic of Korea
| | - Ye-Eun Lee
- Department of Efficacy Study, Institute of Jinan Red Ginseng, Jinan-gun, Jeollabuk-do, Republic of Korea
| | - Hyo-Bin Oh
- Department of Efficacy Study, Institute of Jinan Red Ginseng, Jinan-gun, Jeollabuk-do, Republic of Korea
- Department of Food Science and Technology, Jeonbuk National University, Deokjin-gu, Jeonju-si, Jeollabuk-do, Republic of Korea
| | - Chung-berm Park
- Department of Efficacy Study, Institute of Jinan Red Ginseng, Jinan-gun, Jeollabuk-do, Republic of Korea
- Corresponding author. Institute of Jinan Red Ginseng, 41 Hongsamhanbang-ro, Jinan-gun, Jeollabuk-do, 55442, Republic of Korea.
| | - Bumseok Kim
- Biosafety Research Institute and Laboratory of Pathology, College of Veterinary Medicine, Jeonbuk National University, Iksan-si, Jeollabuk-do, Republic of Korea
- Corresponding author. Biosafety Research Institute and Laboratory of Pathology, College of Veterinary Medicine, Jeonbuk National University, 79 Gobong-ro, Iksan-si, Jeollabukdo, 54596, Republic of Korea.
| |
Collapse
|
44
|
Li W, Shen Y, Gong X, Zhang XB, Yuan L. Highly Selective Fluorescent Probe Design for Visualizing Hepatic Hydrogen Sulfide in the Pathological Progression of Nonalcoholic Fatty Liver. Anal Chem 2021; 93:16673-16682. [PMID: 34842411 DOI: 10.1021/acs.analchem.1c04246] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Hydrogen sulfide (H2S), emerging as an important gaseous signal, has attracted more and more attention for its key role in chronic fatty liver diseases. However, lacking tools for H2S-specific in situ detection, the changes of endogenous hepatic H2S levels in the pathological progression of chronic liver diseases are still unclear. To this end, we adopted a strategy of combining molecular probe design and nanofunctionalization to develop a highly selective near-infrared (NIR) fluorescent probe, which allows in vivo real-time monitoring of hepatic H2S levels in the process of nonalcoholic fatty liver disease (NAFLD). As a proof of strategy demonstration, we first designed NIR molecular probes for H2S sensing through chemical design and probe screening and then loaded molecular probes into mesoporous silicon nanomaterials (MSNs) with surface encapsulation using poly(ethylene glycol) to construct a highly selective probe MSN@CSN@PEG, with significantly improved selectivity and photostability. Moreover, MSN@CSN@PEG exhibited high selectivity and sensitivity for endogenous H2S in cells and tumors in vivo, eliminating the interference of a high concentration of biothiols and sulfhydryl proteins. Furthermore, the probe was applied to in situ intravital imaging and systematic assessment of hepatic H2S levels in different stages of NAFLD for the first time, which may offer a promising tool for the future study of fatty liver diseases and other chronic liver diseases.
Collapse
Affiliation(s)
- Wei Li
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Yang Shen
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Xiangyang Gong
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Xiao-Bing Zhang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Lin Yuan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| |
Collapse
|
45
|
The Effects of Butyrate on Induced Metabolic-Associated Fatty Liver Disease in Precision-Cut Liver Slices. Nutrients 2021; 13:nu13124203. [PMID: 34959755 PMCID: PMC8703944 DOI: 10.3390/nu13124203] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/13/2021] [Accepted: 11/23/2021] [Indexed: 02/06/2023] Open
Abstract
Metabolic-associated fatty liver disease (MAFLD) starts with hepatic triglyceride accumulation (steatosis) and can progress to more severe stages such as non-alcoholic steatohepatitis (NASH) and even cirrhosis. Butyrate, and butyrate-producing bacteria, have been suggested to reduce liver steatosis directly and systemically by increasing liver β-oxidation. This study aimed to examine the influence of butyrate directly on the liver in an ex vivo induced MAFLD model. To maintain essential intercellular interactions, precision-cut liver slices (PCLSs) were used. These PCLSs were prepared from male C57BL/6J mice and cultured in varying concentrations of fructose, insulin, palmitic acid and oleic acid, to mimic metabolic syndrome. Dose-dependent triglyceride accumulation was measured after 24 and 48 h of incubation with the different medium compositions. PCLSs viability, as indicated by ATP content, was not affected by medium composition or the butyrate concentration used. Under induced steatotic conditions, butyrate did not prevent triglyceride accumulation. Moreover, it lowered the expression of genes encoding for fatty acid oxidation and only increased C4 related carnitines, which indicate butyrate oxidation. Nevertheless, butyrate lowered the fibrotic response of PCLSs, as shown by reduced gene expression of fibronectin, alpha-smooth muscle actin and osteopontin, and protein levels of type I collagen. These results suggest that in the liver, butyrate alone does not increase lipid β-oxidation directly but might aid in the prevention of MAFLD progression to NASH and cirrhosis.
Collapse
|
46
|
Obydah WO, Shaker GA, Samir SM, El Bassiony SF, Abd El Moneim HA. Effect of vanillic acid and exercise training on fatty liver and insulin resistance in rats: Possible role of fibroblast growth factor 21 and autophagy. Physiol Int 2021; 108:412-426. [PMID: 34813496 DOI: 10.1556/2060.2021.00188] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 10/22/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND AND AIMS The prevalence of non-alcoholic fatty liver disease has been alarmingly increased with no lines of effective treatment. Vanillic acid is a naturally occurring polyphenol with promising therapeutic effects. Exercise is well known to be an effective tool against obesity and its consequences. Thus, we aim to study the effect of vanillic acid alone and along with exercise on fatty liver induced by a high-fat diet in a rat model and to investigate possible novel mechanisms involved in their action. METHODS In this study, 40 male rats were divided equally into five groups: control (standard chow diet), HFD (high-fat diet), HFD+VA (HFD+ vanillic acid (50 mg/kg/day orally), HFD+EX (HFD+ swimming exercise 5 days/week), HFD+VA+EX (HFD+ vanillic acid+ swimming exercise) for eight weeks. RESULTS Body mass, liver weight, liver enzymes, cholesterol, and triglycerides were significantly decreased in the combined VA+EX group, with marked improvement in hyperglycemia, hyperinsulinemia, and consequently HOMA-IR index compared to the HFD group. These improvements were also reflected in the pathological view. VA and swimming, either solely or in combination, markedly increased hepatic and circulating fibroblast growth factor 21. Additionally, VA and swimming increased the immunohistochemical expression of the autophagosomal marker LC3 and decreased the expression of P62, which is selectively degraded during autophagy. CONCLUSIONS These results suggest the hepatoprotective effect of VA and swimming exercise against fatty liver and the involvement of FGF21 and autophagy in their effect.
Collapse
Affiliation(s)
- Walaa O Obydah
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Egypt
| | - Gehan A Shaker
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Egypt
| | - Shereen M Samir
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Egypt
| | - Soheir F El Bassiony
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Egypt
| | | |
Collapse
|
47
|
Anti-Obesity Effects of Matoa ( Pometia pinnata) Fruit Peel Powder in High-Fat Diet-Fed Rats. Molecules 2021; 26:molecules26216733. [PMID: 34771142 PMCID: PMC8588254 DOI: 10.3390/molecules26216733] [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: 10/11/2021] [Revised: 11/03/2021] [Accepted: 11/03/2021] [Indexed: 12/21/2022] Open
Abstract
Fruit peels, pericarps, or rinds are rich in phenolic/polyphenolic compounds with antioxidant properties and potentially beneficial effects against obesity and obesity-related non-communicable diseases. This study investigated the anti-obesity effects of matoa (Pometia pinnata) and salak (Salacca zalacca) fruit peel. Neither matoa peel powder (MPP) nor salak peel powder (SPP) affected the body weight, visceral fat weight, or serum glucose or lipid levels of Sprague-Dawley rats when included as 1% (w/w) of a high-fat diet (HFD). However, MPP significantly decreased the hepatic lipid level. MPP at a dose of 3% (w/w) of the HFD decreased body weight, visceral fat, and serum triglyceride levels as well as the hepatic lipid content. The inhibitory effect of MPP on hepatic lipid accumulation was not enhanced when its concentration was increased from 1% to 3% of the HFD. The anti-obesity effect of matoa was partly explained by the inhibitory effect of the matoa peel extract on fatty acid-induced secretion of ApoB-48 protein, a marker of intestinal chylomicrons, in differentiated Caco-2 cell monolayers. We identified hederagenin saponins that are abundant in MPP as potential anti-obesity substances. These results will contribute towards the development of functional foods with anti-obesity effects using the matoa fruit peel.
Collapse
|
48
|
SREBP1c silencing reduces endoplasmic reticulum stress and related apoptosis in oleic acid induced lipid accumulation. MARMARA MEDICAL JOURNAL 2021. [DOI: 10.5472/marumj.1009096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
49
|
An Efficient Model of Non-alcoholic Fatty Liver Disease (NAFLD) Versus Current Experimental Models: Effects of Fructose, Fat, and Carbon Tetrachloride on NAFLD. HEPATITIS MONTHLY 2021. [DOI: 10.5812/hepatmon.117696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Abstract
Background: Accumulation of fat in the liver is one of the causes of non-alcoholic fatty liver disease (NAFLD), which affects about 30% of the world's population. Animal models have been useful tools for investigating the mechanisms involved in the etiology of NAFLD and developing new drugs. Objectives: This study aimed to present a new model for the detection of NAFLD in rats. Methods: Forty-eight rats were randomly divided into six experimental groups: (1) control; (2) 45% fructose + 35% olive oil + carbon tetrachloride (FFC1); (3) carbon tetrachloride (1: 4 in olive oil) (C1); (4) carbon tetrachloride (1: 6 in olive oil) (C2); (5) 12.5% fructose + 12.5% olive oil (FF); and (6) 20% fructose + carbon tetrachloride (1: 4 in olive oil) (FC1). Blood samples were taken in three steps, and liver tissue was dissected at the end of the sixth week for histopathological assessments. Results: After six weeks, the alanine transaminase (131.63 ± 1.51), aspartate transaminase (275 ± 1.0), and gamma-glutamyl transferase (4.30 ± 0.1) levels increased significantly in the C1 group (P < 0.05). The serum lipid profile showed significant changes in all groups compared to the controls (P < 0.01). According to the histological results, all experimental groups, except the C2 group, showed symptoms of NAFLD; nevertheless, a higher NAFLD Activity Score (NAS) was found in the C1 group, followed by the FC1 group, compared to the other groups. Conclusions: The present results revealed that injection of 0.1 mL/kg of carbon tetrachloride (C1 group), alone or along with a diet containing 20% fructose (FC1 group), provided useful animal models of NAFLD, although carbon tetrachloride injection alone is the most effective model in inducing NAFLD model that can be used as a new strategy in nutritional and pharmacological studies.
Collapse
|
50
|
Antifibrotic Effects of Kangxian Ruangan Capsule on Rats with Nonalcoholic Fatty Liver Fibrosis and Hepatic Stellate Cells through Regulation of TGF- β and TLR4 Signaling Pathways. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:5649575. [PMID: 34422075 PMCID: PMC8371615 DOI: 10.1155/2021/5649575] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 05/07/2021] [Accepted: 07/22/2021] [Indexed: 01/18/2023]
Abstract
Kangxian ruangan (KXRG) is a traditional Chinese medicine (TCM) formula consisting of 12 herbs. TCM syndrome differentiation proposes that KXRG exerts pharmacological effects against nonalcoholic fatty liver disease (NAFLD) fibrosis. This work investigates the effect of KXRG on NAFLD fibrosis in vivo and in vitro. In vivo, the NAFLD fibrosis model was constructed in Wistar rats using methionine- and choline-deficient (MCD) diet, followed by KXRG (0.92 g/kg/d) treatment for 8 weeks. In vitro, primary hepatic stellate cells (HSCs) were activated using platelet-derived growth factor (PDGF) and treated with KXRG. Molecular mechanisms underlying fibrosis were investigated. After 8 weeks, compared with the control groups, the histological lesions, degree of fibrosis, and inflammatory reaction increased with the MCD diet as demonstrated by histological changes and increased fibrosis-related (α-SMA, TGF-β, COL1A1, and desmin, P < 0.01) and inflammation-related factors (TNF-α, MCP-1, and F4/80, P < 0.01), whereas they decreased with KXRG treatment (P < 0.01). KXRG not only inhibited the proliferation of activated HSCs and promoted their apoptosis but also resulted in G0-G1 arrest. Furthermore, KXRG suppressed HSC activation (P < 0.01), collagen synthesis (P < 0.01), and α-SMA expression (P < 0.01) with PDGF stimulation. In both the MCD diet-induced animal model and PDGF-induced cell model, KXRG inhibited TGF-β and TLR4 signaling (P < 0.01), similar to corresponding small-molecule inhibitors. These results demonstrated that KXRG might exert suppressive effects against NAFLD fibrosis via regulating TGF-β and TLR4 signaling. KXRG may act as a natural and potent therapeutic agent against NAFLD.
Collapse
|