1
|
Zhang M, Ji J, Lei Y, Qin F, Tao Y, Li N, Bian J, Li Z, Lai M, Qiu Z. Dual inhibition of hepatic ACLY and ACSS2: A synergistic approach to combat NAFLD through lipogenesis reduction and mitochondrial enhancement. Pharmacol Res 2025; 215:107706. [PMID: 40127788 DOI: 10.1016/j.phrs.2025.107706] [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: 01/14/2025] [Revised: 02/28/2025] [Accepted: 03/18/2025] [Indexed: 03/26/2025]
Abstract
Inhibiting de novo lipogenesis (DNL) in hepatocytes is a promising strategy for treating metabolic fatty liver diseases. ACLY, a key enzyme in the DNL pathway, has become a therapeutic target for non-alcoholic fatty liver disease (NAFLD). However, its inhibition shows mixed outcomes, depending on interventions and diets. Evidence suggests ACLY inhibition activates the ACSS2-mediated acetate metabolism and the subsequent DNL, though potential mechanisms and possible consequences remain unclear. This study found that targeting hepatic ACLY with AAV8-shRNA failed to improve NAFLD in mice fed a high-fat, high-fructose diet. Instead, it worsened inflammation and liver injury. ACLY inhibition conditionally upregulated DNL enzymes, but consistently activated the ACSS2-acetyl-CoA pathway and suppressed fatty acid oxidation. Further, ACLY inhibition led to polyunsaturated fatty acid accumulation, triggering mitochondrial dysfunction. The resulting ROS redirected carbon flux into acetate, activating the ACSS2-acetyl-CoA pathway, which promoted lipid biosynthesis and exacerbated mitochondrial dysfunction-a vicious cycle that fueled inflammation and liver damage. Dual inhibition of ACLY and ACSS2 broke this cycle by reducing hepatic acetyl-CoA flux, suppressing DNL, enhancing fatty acid oxidation via PPAR-α activation, and improving mitochondrial function. This combined targeting strategy reduced lipid accumulation, alleviated inflammation, and normalized aminotransferase levels, effectively reversing NAFLD progression.
Collapse
Affiliation(s)
- Mengdi Zhang
- School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jinliang Ji
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yuanyuan Lei
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Fujian Qin
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yitong Tao
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Ning Li
- National Experimental Teaching Demonstration Center of Pharmacy, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jinlei Bian
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Zhiyu Li
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Maode Lai
- School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Disease Proteomics of Zhejiang Province, Department of Pathology, Zhejiang University School of Medicine, Hangzhou 310058, China; Research Unit of Intelligence Classification of Tumor Pathology and Precision Therapy, Chinese Academy of Medical Sciences (2019RU042), Department of Pathology, Zhejiang University School of Medicine, Hangzhou 310058, China.
| | - Zhixia Qiu
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
2
|
Khayatan D, Razavi SM, Arab ZN, Nasoori H, Fouladi A, Pasha AVK, Butler AE, Karav S, Momtaz S, Abdolghaffari AH, Sahebkar A. Targeting mTOR with curcumin: therapeutic implications for complex diseases. Inflammopharmacology 2025; 33:1583-1616. [PMID: 39955697 DOI: 10.1007/s10787-025-01643-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/16/2024] [Indexed: 02/17/2025]
Abstract
The mammalian target of rapamycin (mTOR) is a crucial enzyme in regulating multiple signaling pathways in the body, including autophagy, proliferation and apoptosis. Disruption of these mTOR signaling pathways can lead to an array of abnormalities and trigger disease processes, examples being neurodegenerative conditions, cancer, obesity and diabetes. Under conditions of oxidative stress, mTOR can regulate apoptosis and autophagy, with tissue repair being favored under such circumstances. Moreover, the correlation between mTOR and other signaling pathways could play a pivotal role in the pathophysiology of numerous disorders. mTOR has a tight connection with NF-κB, Akt, PI3K, MAPK, GSK-3β, Nrf2/HO-1, JAK/STAT, CREB/BDNF, and ERK1/2 pathways, which together could play significant roles in the regulation of inflammation, apoptosis, cell survival, and oxidative stress in different body organs. Research suggests that inhibiting mTOR could be beneficial in treating metabolic, neurological and cardiovascular conditions, as well as potentially extending life expectancy. Therefore, identifying new chemicals and agents that can modulate the mTOR signaling pathway holds promise for treating and preventing these disorders. Curcumin is one such agent that has demonstrated regulatory effects on the mTOR pathway, making it an exciting alternative for reducing complications associated with complex diseases by targeting mTOR. This review aims to examine the potential of curcumin in modulating the mTOR signaling pathway and its therapeutic implications.
Collapse
Affiliation(s)
- Danial Khayatan
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Seyed Mehrad Razavi
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Zahra Najafi Arab
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hadis Nasoori
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Abtin Fouladi
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Aytak Vahdat Khajeh Pasha
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland, Bahrain, Adliya, Bahrain
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Canakkale Onsekiz Mart University, Canakkale, 17100, Turkey
| | - Saeideh Momtaz
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran
- Department of Toxicology and Pharmacology, School of Pharmacy, and Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Hossein Abdolghaffari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Zhang Y, Yang J, Min J, Huang S, Li Y, Liu S. The emerging role of E3 ubiquitin ligases and deubiquitinases in metabolic dysfunction-associated steatotic liver disease. J Transl Med 2025; 23:368. [PMID: 40133964 PMCID: PMC11938720 DOI: 10.1186/s12967-025-06255-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 02/17/2025] [Indexed: 03/27/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common chronic liver disease worldwide, with a prevalence as high as 32.4%. MASLD encompasses a spectrum of liver pathologies, ranging from steatosis to metabolic dysfunction-associated steatohepatitis (MASH), fibrosis, and, in some cases, progression to end-stage liver disease (cirrhosis and hepatocellular carcinoma). A comprehensive understanding of the pathogenesis of this highly prevalent liver disease may facilitate the identification of novel targets for the development of improved therapies. E3 ubiquitin ligases and deubiquitinases (DUBs) are key regulatory components of the ubiquitin‒proteasome system (UPS), which plays a pivotal role in maintaining intracellular protein homeostasis. Emerging evidence implicates that aberrant expression of E3 ligases and DUBs is involved in the progression of MASLD. Here, we review abnormalities in E3 ligases and DUBs by (1) discussing their targets, mechanisms, and functions in MASLD; (2) summarizing pharmacological interventions targeting these enzymes in preclinical and clinical studies; and (3) addressing challenges and future therapeutic strategies. This review synthesizes current evidence to highlight the development of novel therapeutic strategies based on the UPS for MASLD and progressive liver disease.
Collapse
Affiliation(s)
- Yu Zhang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, CSU-Sinocare Research Center for Nutrition and Metabolic Health, Furong Laboratory, Changsha, Hunan, 410011, China
| | - Jiahui Yang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, CSU-Sinocare Research Center for Nutrition and Metabolic Health, Furong Laboratory, Changsha, Hunan, 410011, China
| | - Jiali Min
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, CSU-Sinocare Research Center for Nutrition and Metabolic Health, Furong Laboratory, Changsha, Hunan, 410011, China
| | - Shan Huang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, CSU-Sinocare Research Center for Nutrition and Metabolic Health, Furong Laboratory, Changsha, Hunan, 410011, China
| | - Yuchen Li
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, CSU-Sinocare Research Center for Nutrition and Metabolic Health, Furong Laboratory, Changsha, Hunan, 410011, China
| | - Shanshan Liu
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, CSU-Sinocare Research Center for Nutrition and Metabolic Health, Furong Laboratory, Changsha, Hunan, 410011, China.
| |
Collapse
|
4
|
Sabini JH, Timotius KH. Hepatoprotective and Fat-Accumulation-Reductive Effects of Curcumin on Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD). Curr Issues Mol Biol 2025; 47:159. [PMID: 40136412 PMCID: PMC11940900 DOI: 10.3390/cimb47030159] [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: 01/16/2025] [Revised: 02/06/2025] [Accepted: 02/25/2025] [Indexed: 03/27/2025] Open
Abstract
Fat accumulation is the hallmark of metabolic dysfunction-associated steatotic liver disease (MASLD). Given the intimidating nature of its treatment, curcumin (CUR) emerges as a potential therapeutic agent due to its proven effectiveness in managing MASLD. This review aimed to evaluate previous reports on the hepatoprotective and fat-accumulation-reductive effects of CUR administration in preventing or treating MASLD. CUR administration can modulate serum liver enzymes and lipid profiles. The fat accumulation of MASLD is the primary cause of oxidative stress and inflammation. By reducing fat accumulation, CUR may attenuate the inflammation and oxidative stress in MASLD. In addition, CUR has been proven to restore the dysfunctional cellular energy metabolism capacity and attenuate fibrogenesis (antifibrotic agent). Their hepatoprotective effects are associated with fat accumulation in MASLD. Lipid metabolism (lipogenesis, lipolysis, and lipophagy) is correlated with their hepatoprotective effects. CUR has prophylactic and therapeutic effects, particularly in early-stage MASLD, primarily when it is used as a fat reducer. It can be considered an excellent natural therapeutic drug for MASLD because it protects the liver and attenuates fat accumulation, especially in the early stage of MASLD development.
Collapse
Affiliation(s)
| | - Kris Herawan Timotius
- Faculty of Medicine and Health Sciences, Krida Wacana Christian University, Jakarta 11510, Indonesia;
| |
Collapse
|
5
|
Cai Y, Fang L, Chen F, Zhong P, Zheng X, Xing H, Fan R, Yuan L, Peng W, Li X. Targeting AMPK related signaling pathways: A feasible approach for natural herbal medicines to intervene non-alcoholic fatty liver disease. J Pharm Anal 2025; 15:101052. [PMID: 40034684 PMCID: PMC11873010 DOI: 10.1016/j.jpha.2024.101052] [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: 12/03/2023] [Revised: 07/13/2024] [Accepted: 07/22/2024] [Indexed: 03/05/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a metabolic disease characterized by abnormal deposition of lipid in hepatocytes. If not intervened in time, NAFLD may develop into liver fibrosis or liver cancer, and ultimately threatening life. NAFLD has complicated etiology and pathogenesis, and there are no effective therapeutic means and specific drugs. Currently, insulin sensitizers, lipid-lowering agents and hepatoprotective agents are often used for clinical intervention, but these drugs have obvious side effects, and their effectiveness and safety need to be further confirmed. Adenosine monophosphate (AMP)-activated protein kinase (AMPK) plays a central role in maintaining energy homeostasis. Activated AMPK can enhance lipid degradation, alleviate insulin resistance (IR), suppress oxidative stress and inflammatory response, and regulate autophagy, thereby alleviating NAFLD. Natural herbal medicines have received extensive attention recently because of their regulatory effects on AMPK and low side effects. In this article, we reviewed the biologically active natural herbal medicines (such as natural herbal medicine formulas, extracts, polysaccharides, and monomers) that reported in recent years to treat NAFLD via regulating AMPK, which can serve as a foundation for subsequent development of candidate drugs for NAFLD.
Collapse
Affiliation(s)
- Yongqing Cai
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Lu Fang
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing, 400016, China
| | - Fei Chen
- Department of Pharmacy, Dazhou Integrated Traditional Chinese Medicine and Western Medicine Hospital, Dazhou, Sichuan, 635000, China
| | - Peiling Zhong
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing, 400016, China
| | - Xiangru Zheng
- Department of Pharmacy, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Haiyan Xing
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Rongrong Fan
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, 14152, Sweden
| | - Lie Yuan
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing, 400016, China
| | - Wei Peng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Xiaoli Li
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing, 400016, China
| |
Collapse
|
6
|
Mo L, Wan S, Zékány-Nagy T, Luo X, Yang X. The Effect of Curcumin on Glucolipid Metabolic Disorders: A Review. FOOD REVIEWS INTERNATIONAL 2024:1-35. [DOI: 10.1080/87559129.2024.2405654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Affiliation(s)
- Lifen Mo
- Food Safety and Health Research Center, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong-Hongkong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, PR China
| | - Siyu Wan
- Food Safety and Health Research Center, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong-Hongkong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, PR China
| | - Tekla Zékány-Nagy
- Food Safety and Health Research Center, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong-Hongkong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, PR China
| | - Xiaoyi Luo
- Food Safety and Health Research Center, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong-Hongkong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, PR China
| | - Xingfen Yang
- Food Safety and Health Research Center, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong-Hongkong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, PR China
| |
Collapse
|
7
|
Chen H, Zhou Y, Hao H, Xiong J. Emerging mechanisms of non-alcoholic steatohepatitis and novel drug therapies. Chin J Nat Med 2024; 22:724-745. [PMID: 39197963 DOI: 10.1016/s1875-5364(24)60690-4] [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: 07/02/2024] [Indexed: 09/01/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become a leading cause of chronic liver disease globally. It initiates with simple steatosis (NAFL) and can progress to the more severe condition of non-alcoholic steatohepatitis (NASH). NASH often advances to end-stage liver diseases such as liver fibrosis, cirrhosis, and hepatocellular carcinoma (HCC). Notably, the transition from NASH to end-stage liver diseases is irreversible, and the precise mechanisms driving this progression are not yet fully understood. Consequently, there is a critical need for the development of effective therapies to arrest or reverse this progression. This review provides a comprehensive overview of the pathogenesis of NASH, examines the current therapeutic targets and pharmacological treatments, and offers insights for future drug discovery and development strategies for NASH therapy.
Collapse
Affiliation(s)
- Hao Chen
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yang Zhou
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Haiping Hao
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Jing Xiong
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
8
|
Zhang L, Hu W, Guo H, Sun Q, Xu X, Li Z, Qiu Z, Bian J. Discovery of Highly Potent Solute Carrier 13 Member 5 (SLC13A5) Inhibitors for the Treatment of Hyperlipidemia. J Med Chem 2024; 67:6687-6704. [PMID: 38574002 DOI: 10.1021/acs.jmedchem.4c00260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
In the face of escalating metabolic disease prevalence, largely driven by modern lifestyle factors, this study addresses the critical need for novel therapeutic approaches. We have identified the sodium-coupled citrate transporter (NaCT or SLC13A5) as a target for intervention. Utilizing rational drug design, we developed a new class of SLC13A5 inhibitors, anchored by the hydroxysuccinic acid scaffold, refining the structure of PF-06649298. Among these, LBA-3 emerged as a standout compound, exhibiting remarkable potency with an IC50 value of 67 nM, significantly improving upon PF-06649298. In vitro assays demonstrated LBA-3's efficacy in reducing triglyceride levels in OPA-induced HepG2 cells. Moreover, LBA-3 displayed superior pharmacokinetic properties and effectively lowered triglyceride and total cholesterol levels in diverse mouse models (PCN-stimulated and starvation-induced), without detectable toxicity. These findings not only spotlight LBA-3 as a promising candidate for hyperlipidemia treatment but also exemplify the potential of targeted molecular design in advancing metabolic disorder therapeutics.
Collapse
Affiliation(s)
- Li'ao Zhang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, P. R. China
| | - Wenjun Hu
- Departments of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 211100, P. R. China
| | - Huimin Guo
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, P. R. China
| | - Qiushuang Sun
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, Nanjing 211100, P. R. China
| | - Xi Xu
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, P. R. China
| | - Zhiyu Li
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, P. R. China
| | - Zhixia Qiu
- Departments of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 211100, P. R. China
| | - Jinlei Bian
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, P. R. China
| |
Collapse
|
9
|
Guo Y, Sun Q, Wang S, Zhang M, Lei Y, Wu J, Wang X, Hu W, Meng H, Li Z, Xu L, Huang F, Qiu Z. Corydalis saxicola Bunting total alkaloids improve NAFLD by suppressing de novo lipogenesis through the AMPK-SREBP1 axis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117162. [PMID: 37690477 DOI: 10.1016/j.jep.2023.117162] [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: 07/21/2023] [Revised: 08/24/2023] [Accepted: 09/07/2023] [Indexed: 09/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Along with the gradually increasing incidence, nonalcoholic fatty liver disease (NAFLD) has already been influencing the health of more and more people in the world. Corydalis saxicola Bunting (CSB), a valuable folk medicine, is the dried whole grass of a perennial herb, Yanhuanglian (Papaveraceae), which has significant effects on various hepatitis, liver fibrosis, cirrhosis and other liver diseases. Corydalis saxicola Bunting total alkaloids (CSBTA), a mixture of alkaloids extracted from CSB, exhibit widely-accepted hepatoprotective effects. AIM OF THE STUDY This study aimed to explore the therapeutic potential of CSBTA on NAFLD and the underlying mechanism. MATERIALS AND METHODS A mice model was established by high fat and high cholesterol diet (HFHCD) to study the benefits of CSBTA on the progression of NAFLD. The efficacy of CSBTA on NAFLD was revealed systematically via RNA-sequencing analysis. Further efficacy and molecular mechanism study were explored in mouse primary hepatocytes and HepG2 cells stimulated with high energy with or without pharmacological inhibition or gene silencing. RESULTS CSBTA effectively improved the major hallmarks of NAFLD including liver lipid accumulation, liver injury, inflammation and fibrosis in HFHCD-fed mice. RNA sequencing and targeted qPCR analysis jointly evidenced CSBTA significantly suppressed the expression of Srebf1, Acc1 and Fasn which are the genes responsible for fatty acid biosynthesis. Moreover, stable isotope tracer test denoted CSBTA reduced lipid accumulation via interrupting fatty acid biosynthesis in hepatocytes or the liver. Mechanistically, CSBTA could impede SREBP1 maturation via AMPK activation, thereby reducing DNL-derived lipid accumulation in hepatocytes. CONCLUSIONS CSBTA protected against hepatic steatosis and other hallmarks of NAFLD induced by HFHCD via suppressing DNL, through modulating the AMPK-SREBP1 axis. CSBTA may therefore have a therapeutic potential for NAFLD treatment.
Collapse
Affiliation(s)
- Yating Guo
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Qiushuang Sun
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Shijiao Wang
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Mengdi Zhang
- School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Yuanyuan Lei
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Jiejie Wu
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Xinhong Wang
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Wenjun Hu
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Haitao Meng
- Shimadzu (China) Co., LTD., Nanjing Branch, Nanjing, China.
| | - Zhiyu Li
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Luzhou Xu
- Gastroenterology Department, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.
| | - Fang Huang
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Zhixia Qiu
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
10
|
Li Y, Deng X, Tan X, Li Q, Yu Z, Wu W, Ma X, Zeng J, Wang X. Protective role of curcumin in disease progression from non-alcoholic fatty liver disease to hepatocellular carcinoma: a meta-analysis. Front Pharmacol 2024; 15:1343193. [PMID: 38313314 PMCID: PMC10834658 DOI: 10.3389/fphar.2024.1343193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 01/04/2024] [Indexed: 02/06/2024] Open
Abstract
Background: Pathological progression from non-alcoholic fatty liver disease (NAFLD) to liver fibrosis (LF) to hepatocellular carcinoma (HCC) is a common dynamic state in many patients. Curcumin, a dietary supplement derived from the turmeric family, is expected to specifically inhibit the development of this progression. However, there is a lack of convincing evidence. Methods: The studies published until June 2023 were searched in PubMed, Web of Science, Embase, and the Cochrane Library databases. The SYstematic Review Center for Laboratory animal Experimentation (SYRCLE) approach was used to evaluate the certainty of evidence. StataSE (version 15.1) and Origin 2021 software programs were used to analyze the critical indicators. Results: Fifty-two studies involving 792 animals were included, and three disease models were reported. Curcumin demonstrates a significant improvement in key indicators across the stages of NAFLD, liver fibrosis, and HCC. We conducted a detailed analysis of common inflammatory markers IL-1β, IL-6, and TNF-α, which traverse the entire disease process. The research results reveal that curcumin effectively hinders disease progression at each stage by suppressing inflammation. Curcumin exerted hepatoprotective effects in the dose range from 100 to 400 mg/kg and treatment duration from 4 to 10 weeks. The mechanistic analysis reveals that curcumin primarily exerts its hepatoprotective effects by modulating multiple signaling pathways, including TLR4/NF-κB, Keap1/Nrf2, Bax/Bcl-2/Caspase 3, and TGF-β/Smad3. Conclusion: In summary, curcumin has shown promising therapeutic effects during the overall progression of NAFLD-LF-HCC. It inhibited the pathological progression by synergistic mechanisms related to multiple pathways, including anti-inflammatory, antioxidant, and apoptosis regulation.
Collapse
Affiliation(s)
- Yubing Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xinyu Deng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiyue Tan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qianrong Li
- Department of Obstetrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhi Yu
- Department of Obstetrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wenbin Wu
- Health Care Office of the Service Bureau of Agency for Offices Administration of the Central Military Commission, Beijing, China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinhao Zeng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaoyin Wang
- Department of Obstetrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
11
|
Sun Q, Guo Y, Hu W, Zhang M, Wang S, Lei Y, Meng H, Li N, Xu P, Li Z, Lin H, Huang F, Qiu Z. Bempedoic Acid Unveils Therapeutic Potential in Non-Alcoholic Fatty Liver Disease: Suppression of the Hepatic PXR-SLC13A5/ACLY Signaling Axis. Drug Metab Dispos 2023; 51:1628-1641. [PMID: 37684055 DOI: 10.1124/dmd.123.001449] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 08/25/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023] Open
Abstract
The hepatic SLC13A5/SLC25A1-ATP-dependent citrate lyase (ACLY) signaling pathway, responsible for maintaining the citrate homeostasis, plays a crucial role in the pathogenesis of non-alcoholic fatty liver disease (NAFLD). Bempedoic acid (BA), an ACLY inhibitor commonly used for managing hypercholesterolemia, has shown promising results in addressing hepatic steatosis. This study aimed to elucidate the intricate relationships in processes of hepatic lipogenesis among SLC13A5, SLC25A1, and ACLY and to examine the therapeutic potential of BA in NAFLD, providing insights into its underlying mechanism. In murine primary hepatocytes and HepG2 cells, the silencing or pharmacological inhibition of SLC25A1/ACLY resulted in significant upregulation of SLC13A5 transcription and activity. This increase in SLC13A5 activity subsequently led to enhanced lipogenesis, indicating a compensatory role of SLC13A5 when the SLC25A1/ACLY pathway was inhibited. However, BA effectively counteracted this upregulation, reduced lipid accumulation, and ameliorated various biomarkers of NAFLD. The disease-modifying effects of BA were further confirmed in NAFLD mice. Mechanistic investigations revealed that BA could reverse the elevated transcription levels of SLC13A5 and ACLY, and the subsequent lipogenesis induced by PXR activation in vitro and in vivo. Importantly, this effect was diminished when PXR was knocked down, suggesting the involvement of the hepatic PXR-SLC13A5/ACLY signaling axis in the mechanism of BA action. In conclusion, SLC13A5-mediated extracellular citrate influx emerges as an alternative pathway to SLC25A1/ACLY in the regulation of lipogenesis in hepatocytes, BA exhibits therapeutic potential in NAFLD by suppressing the hepatic PXR-SLC13A5/ACLY signaling axis, while PXR, a key regulator in drug metabolism may be involved in the pathogenesis of NAFLD. SIGNIFICANCE STATEMENT: This work describes that bempedoic acid, an ATP-dependent citrate lyase (ACLY) inhibitor, ameliorates hepatic lipid accumulation and various hallmarks of non-alcoholic fatty liver disease. Suppression of hepatic SLC25A1-ACLY pathway upregulates SLC13A5 transcription, which in turn activates extracellular citrate influx and the subsequent DNL. Whereas in hepatocytes or the liver tissue challenged with high energy intake, bempedoic acid reverses compensatory activation of SLC13A5 via modulating the hepatic PXR-SLC13A5/ACLY axis, thereby simultaneously downregulating SLC13A5 and ACLY.
Collapse
Affiliation(s)
- Qiushuang Sun
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy (Q.S., Y.G., F.H.), Departments of Pharmacology (W.H., S.W., Y.L., Z.Q.) and Medicinal Chemistry, School of Pharmacy (P.X., Z.L.), School of Basic Medical Sciences and Clinical Pharmacy (M.Z.), and National Experimental Teaching Demonstration Center of Pharmacy, School of Pharmacy (N.L.), China Pharmaceutical University, Nanjing, China; Shimadzu (China) Co., LTD., Nanjing Branch, Nanjing, China (H.M.); and College of Pharmacy, Shenzhen Technology University, Shenzhen, China (H.L.)
| | - Yating Guo
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy (Q.S., Y.G., F.H.), Departments of Pharmacology (W.H., S.W., Y.L., Z.Q.) and Medicinal Chemistry, School of Pharmacy (P.X., Z.L.), School of Basic Medical Sciences and Clinical Pharmacy (M.Z.), and National Experimental Teaching Demonstration Center of Pharmacy, School of Pharmacy (N.L.), China Pharmaceutical University, Nanjing, China; Shimadzu (China) Co., LTD., Nanjing Branch, Nanjing, China (H.M.); and College of Pharmacy, Shenzhen Technology University, Shenzhen, China (H.L.)
| | - Wenjun Hu
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy (Q.S., Y.G., F.H.), Departments of Pharmacology (W.H., S.W., Y.L., Z.Q.) and Medicinal Chemistry, School of Pharmacy (P.X., Z.L.), School of Basic Medical Sciences and Clinical Pharmacy (M.Z.), and National Experimental Teaching Demonstration Center of Pharmacy, School of Pharmacy (N.L.), China Pharmaceutical University, Nanjing, China; Shimadzu (China) Co., LTD., Nanjing Branch, Nanjing, China (H.M.); and College of Pharmacy, Shenzhen Technology University, Shenzhen, China (H.L.)
| | - Mengdi Zhang
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy (Q.S., Y.G., F.H.), Departments of Pharmacology (W.H., S.W., Y.L., Z.Q.) and Medicinal Chemistry, School of Pharmacy (P.X., Z.L.), School of Basic Medical Sciences and Clinical Pharmacy (M.Z.), and National Experimental Teaching Demonstration Center of Pharmacy, School of Pharmacy (N.L.), China Pharmaceutical University, Nanjing, China; Shimadzu (China) Co., LTD., Nanjing Branch, Nanjing, China (H.M.); and College of Pharmacy, Shenzhen Technology University, Shenzhen, China (H.L.)
| | - Shijiao Wang
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy (Q.S., Y.G., F.H.), Departments of Pharmacology (W.H., S.W., Y.L., Z.Q.) and Medicinal Chemistry, School of Pharmacy (P.X., Z.L.), School of Basic Medical Sciences and Clinical Pharmacy (M.Z.), and National Experimental Teaching Demonstration Center of Pharmacy, School of Pharmacy (N.L.), China Pharmaceutical University, Nanjing, China; Shimadzu (China) Co., LTD., Nanjing Branch, Nanjing, China (H.M.); and College of Pharmacy, Shenzhen Technology University, Shenzhen, China (H.L.)
| | - Yuanyuan Lei
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy (Q.S., Y.G., F.H.), Departments of Pharmacology (W.H., S.W., Y.L., Z.Q.) and Medicinal Chemistry, School of Pharmacy (P.X., Z.L.), School of Basic Medical Sciences and Clinical Pharmacy (M.Z.), and National Experimental Teaching Demonstration Center of Pharmacy, School of Pharmacy (N.L.), China Pharmaceutical University, Nanjing, China; Shimadzu (China) Co., LTD., Nanjing Branch, Nanjing, China (H.M.); and College of Pharmacy, Shenzhen Technology University, Shenzhen, China (H.L.)
| | - Haitao Meng
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy (Q.S., Y.G., F.H.), Departments of Pharmacology (W.H., S.W., Y.L., Z.Q.) and Medicinal Chemistry, School of Pharmacy (P.X., Z.L.), School of Basic Medical Sciences and Clinical Pharmacy (M.Z.), and National Experimental Teaching Demonstration Center of Pharmacy, School of Pharmacy (N.L.), China Pharmaceutical University, Nanjing, China; Shimadzu (China) Co., LTD., Nanjing Branch, Nanjing, China (H.M.); and College of Pharmacy, Shenzhen Technology University, Shenzhen, China (H.L.)
| | - Ning Li
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy (Q.S., Y.G., F.H.), Departments of Pharmacology (W.H., S.W., Y.L., Z.Q.) and Medicinal Chemistry, School of Pharmacy (P.X., Z.L.), School of Basic Medical Sciences and Clinical Pharmacy (M.Z.), and National Experimental Teaching Demonstration Center of Pharmacy, School of Pharmacy (N.L.), China Pharmaceutical University, Nanjing, China; Shimadzu (China) Co., LTD., Nanjing Branch, Nanjing, China (H.M.); and College of Pharmacy, Shenzhen Technology University, Shenzhen, China (H.L.)
| | - Pengfei Xu
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy (Q.S., Y.G., F.H.), Departments of Pharmacology (W.H., S.W., Y.L., Z.Q.) and Medicinal Chemistry, School of Pharmacy (P.X., Z.L.), School of Basic Medical Sciences and Clinical Pharmacy (M.Z.), and National Experimental Teaching Demonstration Center of Pharmacy, School of Pharmacy (N.L.), China Pharmaceutical University, Nanjing, China; Shimadzu (China) Co., LTD., Nanjing Branch, Nanjing, China (H.M.); and College of Pharmacy, Shenzhen Technology University, Shenzhen, China (H.L.)
| | - Zhiyu Li
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy (Q.S., Y.G., F.H.), Departments of Pharmacology (W.H., S.W., Y.L., Z.Q.) and Medicinal Chemistry, School of Pharmacy (P.X., Z.L.), School of Basic Medical Sciences and Clinical Pharmacy (M.Z.), and National Experimental Teaching Demonstration Center of Pharmacy, School of Pharmacy (N.L.), China Pharmaceutical University, Nanjing, China; Shimadzu (China) Co., LTD., Nanjing Branch, Nanjing, China (H.M.); and College of Pharmacy, Shenzhen Technology University, Shenzhen, China (H.L.)
| | - Haishu Lin
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy (Q.S., Y.G., F.H.), Departments of Pharmacology (W.H., S.W., Y.L., Z.Q.) and Medicinal Chemistry, School of Pharmacy (P.X., Z.L.), School of Basic Medical Sciences and Clinical Pharmacy (M.Z.), and National Experimental Teaching Demonstration Center of Pharmacy, School of Pharmacy (N.L.), China Pharmaceutical University, Nanjing, China; Shimadzu (China) Co., LTD., Nanjing Branch, Nanjing, China (H.M.); and College of Pharmacy, Shenzhen Technology University, Shenzhen, China (H.L.)
| | - Fang Huang
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy (Q.S., Y.G., F.H.), Departments of Pharmacology (W.H., S.W., Y.L., Z.Q.) and Medicinal Chemistry, School of Pharmacy (P.X., Z.L.), School of Basic Medical Sciences and Clinical Pharmacy (M.Z.), and National Experimental Teaching Demonstration Center of Pharmacy, School of Pharmacy (N.L.), China Pharmaceutical University, Nanjing, China; Shimadzu (China) Co., LTD., Nanjing Branch, Nanjing, China (H.M.); and College of Pharmacy, Shenzhen Technology University, Shenzhen, China (H.L.)
| | - Zhixia Qiu
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy (Q.S., Y.G., F.H.), Departments of Pharmacology (W.H., S.W., Y.L., Z.Q.) and Medicinal Chemistry, School of Pharmacy (P.X., Z.L.), School of Basic Medical Sciences and Clinical Pharmacy (M.Z.), and National Experimental Teaching Demonstration Center of Pharmacy, School of Pharmacy (N.L.), China Pharmaceutical University, Nanjing, China; Shimadzu (China) Co., LTD., Nanjing Branch, Nanjing, China (H.M.); and College of Pharmacy, Shenzhen Technology University, Shenzhen, China (H.L.)
| |
Collapse
|
12
|
Wang L, Yan Y, Wu L, Peng J. Natural products in non-alcoholic fatty liver disease (NAFLD): Novel lead discovery for drug development. Pharmacol Res 2023; 196:106925. [PMID: 37714392 DOI: 10.1016/j.phrs.2023.106925] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/06/2023] [Accepted: 09/12/2023] [Indexed: 09/17/2023]
Abstract
With changing lifestyles, non-alcoholic fatty liver disease (NAFLD) has become the most prevalent liver disease worldwide. A substantial increase in the incidence, mortality, and associated burden of NAFLD-related advanced liver disease is expected. Currently, the initial diagnosis of NAFLD is still based on ultrasound and there is no approved treatment method. Lipid-lowering drugs, vitamin supplementation, and lifestyle improvement treatments are commonly used in clinical practice. However, most lipid-lowering drugs can produce poor patient compliance and specific adverse effects. Therefore, the exploration of bio-diagnostic markers and active lead compounds for the development of innovative drugs is urgently needed. More and more studies have reported the anti-NAFLD effects and mechanisms of natural products (NPs), which have become an important source for new drug development to treat NAFLD due to their high activity and low side effects. At present, berberine and silymarin have been approved by the US FDA to enter clinical phase IV studies, demonstrating the potential of NPs against NAFLD. Studies have found that the regulation of lipid metabolism, insulin resistance, oxidative stress, and inflammation-related pathways may play important roles in the process. With the continuous updating of technical means and scientific theories, in-depth research on the targets and mechanisms of NPs against NAFLD can provide new possibilities to find bio-diagnostic markers and innovative drugs. As we know, FXR agonists, PPARα agonists, and dual CCR2/5 inhibitors are gradually coming on stage for the treatment of NAFLD. Whether NPs can exert anti-NAFLD effects by regulating these targets or some unknown targets remains to be further studied. Therefore, the study reviewed the potential anti-NAFLD NPs and their targets. Some works on the discovery of new targets and the docking of active lead compounds were also discussed. It is hoped that this review can provide some reference values for the development of non-invasive diagnostic markers and new drugs against NAFLD in the clinic.
Collapse
Affiliation(s)
- Lu Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Yonghuan Yan
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Linfang Wu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Jinyong Peng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China.
| |
Collapse
|
13
|
Fang X, Song J, Zhou K, Zi X, Sun B, Bao H, Li L. Molecular Mechanism Pathways of Natural Compounds for the Treatment of Non-Alcoholic Fatty Liver Disease. Molecules 2023; 28:5645. [PMID: 37570615 PMCID: PMC10419790 DOI: 10.3390/molecules28155645] [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: 07/03/2023] [Revised: 07/21/2023] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease in the world, and its incidence continues to increase each year. Yet, there is still no definitive drug that can stop its development. This review focuses mainly on lipotoxicity, oxidative stress, inflammation, and intestinal flora dysbiosis to understand NAFLD's pathogenesis. In this review, we used NCBI's PubMed database for retrieval, integrating in vivo and in vitro experiments to reveal the therapeutic effects of natural compounds on NAFLD. We also reviewed the mechanisms by which the results of these experiments suggest that these compounds can protect the liver from damage by modulating inflammation, reducing oxidative stress, decreasing insulin resistance and lipid accumulation in the liver, and interacting with the intestinal microflora. The natural compounds discussed in these papers target a variety of pathways, such as the AMPK pathway and the TGF-β pathway, and have significant therapeutic effects. This review aims to provide new possible therapeutic lead compounds and references for the development of novel medications and the clinical treatment of NAFLD. It offers fresh perspectives on the development of natural compounds in preventing and treating NAFLD.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Lijing Li
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (X.F.)
| |
Collapse
|
14
|
Qin T, Chen X, Meng J, Guo Q, Xu S, Hou S, Yuan Z, Zhang W. The role of curcumin in the liver-gut system diseases: from mechanisms to clinical therapeutic perspective. Crit Rev Food Sci Nutr 2023; 64:8822-8851. [PMID: 37096460 DOI: 10.1080/10408398.2023.2204349] [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] [Indexed: 04/26/2023]
Abstract
Natural products have provided abundant sources of lead compounds for new drug discovery and development over the past centuries. Curcumin is a lipophilic polyphenol isolated from turmeric, a plant used in traditional Asian medicine for centuries. Despite the low oral bioavailability, curcumin exhibits profound medicinal value in various diseases, especially liver and gut diseases, bringing an interest in the paradox of its low bioavailability but high bioactivity. Several latest studies suggest that curcumin's health benefits may rely on its positive gastrointestinal effects rather than its poor bioavailability solely. Microbial antigens, metabolites, and bile acids regulate metabolism and immune responses in the intestine and liver, suggesting the possibility that the liver-gut axis bidirectional crosstalk controls gastrointestinal health and diseases. Accordingly, these pieces of evidence have evoked great interest in the curcumin-mediated crosstalk among liver-gut system diseases. The present study discussed the beneficial effects of curcumin against common liver and gut diseases and explored the underlying molecular targets, as well as collected evidence from human clinical studies. Moreover, this study summarized the roles of curcumin in complex metabolic interactions in liver and intestine diseases supporting the application of curcumin in the liver-gut system as a potential therapeutic option, which opens an avenue for clinical use in the future.
Collapse
Affiliation(s)
- Tingting Qin
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Xiuying Chen
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jiahui Meng
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Qianqian Guo
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Shan Xu
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Shanshan Hou
- Department of Pharmacy, Zhejiang Pharmaceutical College, Ningbo, China
| | - Ziqiao Yuan
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Wenzhou Zhang
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
15
|
Curcumin and Andrographolide Co-Administration Safely Prevent Steatosis Induction and ROS Production in HepG2 Cell Line. Molecules 2023; 28:molecules28031261. [PMID: 36770927 PMCID: PMC9919300 DOI: 10.3390/molecules28031261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/19/2023] [Accepted: 01/24/2023] [Indexed: 02/03/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is an emerging chronic liver disease worldwide. Curcumin and andrographolide are famous for improving hepatic functions, being able to reverse oxidative stress and release pro-inflammatory cytokines, and they are implicated in hepatic stellate cell activation and in liver fibrosis development. Thus, we tested curcumin and andrographolide separately and in combination to determine their effect on triglyceride accumulation and ROS production, identifying the differential expression of genes involved in fatty liver and oxidative stress development. In vitro steatosis was induced in HepG2 cells and the protective effect of curcumin, andrographolide, and their combination was observed evaluating cell viability, lipid and triglyceride content, ROS levels, and microarray differential gene expression. Curcumin, andrographolide, and their association were effective in reducing steatosis, triglyceride content, and ROS stress, downregulating the genes involved in lipid accumulation. Moreover, the treatments were able to protect the cytotoxic effect of steatosis, promoting the expression of survival and anti-inflammatory genes. The present study showed that the association of curcumin and andrographolide could be used as a therapeutic approach to counter high lipid content and ROS levels in steatosis liver, avoiding the possible hepatotoxic effect of curcumin. Furthermore, this study improved our understanding of the antisteatosis and hepatoprotective properties of a curcumin and andrographolide combination.
Collapse
|
16
|
He H, Wang J, Mou X, Liu X, Li Q, Zhong M, Luo B, Yu Z, Zhang J, Xu T, Dou C, Wu D, Qing W, Wu L, Zhou K, Fan Z, Wang T, Hu T, Zhang X, Zhou J, Miao YL. Selective autophagic degradation of ACLY (ATP citrate lyase) maintains citrate homeostasis and promotes oocyte maturation. Autophagy 2023; 19:163-179. [PMID: 35404187 PMCID: PMC9809967 DOI: 10.1080/15548627.2022.2063005] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 04/01/2022] [Accepted: 04/04/2022] [Indexed: 01/07/2023] Open
Abstract
Macroautophagy/autophagy is a cellular and energy homeostatic mechanism that contributes to maintain the number of primordial follicles, germ cell survival, and anti-ovarian aging. However, it remains unknown whether autophagy in granulosa cells affects oocyte maturation. Here, we show a clear tendency of reduced autophagy level in human granulosa cells from women of advanced maternal age, implying a potential negative correlation between autophagy levels and oocyte quality. We therefore established a co-culture system and show that either pharmacological inhibition or genetic ablation of autophagy in granulosa cells negatively affect oocyte quality and fertilization ability. Moreover, our metabolomics analysis indicates that the adverse impact of autophagy impairment on oocyte quality is mediated by downregulated citrate levels, while exogenous supplementation of citrate can significantly restore the oocyte maturation. Mechanistically, we found that ACLY (ATP citrate lyase), which is a crucial enzyme catalyzing the cleavage of citrate, was preferentially associated with K63-linked ubiquitin chains and recognized by the autophagy receptor protein SQSTM1/p62 for selective autophagic degradation. In human follicles, the autophagy level in granulosa cells was downregulated with maternal aging, accompanied by decreased citrate in the follicular fluid, implying a potential correlation between citrate metabolism and oocyte quality. We also show that elevated citrate levels in porcine follicular fluid promote oocyte maturation. Collectively, our data reveal that autophagy in granulosa cells is a beneficial mechanism to maintain a certain degree of citrate by selectively targeting ACLY during oocyte maturation.Abbreviations: 3-MA: 3-methyladenine; ACLY: ATP citrate lyase; AMA: advanced maternal age; CG: cortical granule; CHX: cycloheximide; CQ: chloroquine; CS: citrate synthase; COCs: cumulus-oocyte-complexes; GCM: granulosa cell monolayer; GV: germinal vesicle; MII: metaphase II stage of meiosis; PB1: first polar body; ROS: reactive oxygen species; shRNA: small hairpin RNA; SQSTM1/p62: sequestosome 1; TCA: tricarboxylic acid; TOMM20/TOM20: translocase of outer mitochondrial membrane 20; UBA: ubiquitin-associated domain; Ub: ubiquitin; WT: wild-type.
Collapse
Affiliation(s)
- Hainan He
- Institute of Stem Cell and Regenerative Biology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction (Huazhong Agricultural University), Ministry of Education, Wuhan, Hubei, China
| | - Junling Wang
- Department of Reproductive Medicine, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic, Edong Healthcare Group, Huangshi, Hubei, China
| | - Xingmei Mou
- Institute of Stem Cell and Regenerative Biology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction (Huazhong Agricultural University), Ministry of Education, Wuhan, Hubei, China
| | - Xin Liu
- Institute of Stem Cell and Regenerative Biology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction (Huazhong Agricultural University), Ministry of Education, Wuhan, Hubei, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Qiao Li
- Institute of Stem Cell and Regenerative Biology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction (Huazhong Agricultural University), Ministry of Education, Wuhan, Hubei, China
| | - Mingyue Zhong
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction (Huazhong Agricultural University), Ministry of Education, Wuhan, Hubei, China
| | - Bingbing Luo
- Department of Reproductive Medicine, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic, Edong Healthcare Group, Huangshi, Hubei, China
| | - Zhisheng Yu
- Institute of Stem Cell and Regenerative Biology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction (Huazhong Agricultural University), Ministry of Education, Wuhan, Hubei, China
| | - Jingjing Zhang
- Institute of Stem Cell and Regenerative Biology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction (Huazhong Agricultural University), Ministry of Education, Wuhan, Hubei, China
| | - Tian Xu
- Institute of Stem Cell and Regenerative Biology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction (Huazhong Agricultural University), Ministry of Education, Wuhan, Hubei, China
| | - Chengli Dou
- Institute of Stem Cell and Regenerative Biology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction (Huazhong Agricultural University), Ministry of Education, Wuhan, Hubei, China
| | - Danya Wu
- Institute of Stem Cell and Regenerative Biology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction (Huazhong Agricultural University), Ministry of Education, Wuhan, Hubei, China
| | - Wei Qing
- Institute of Stem Cell and Regenerative Biology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction (Huazhong Agricultural University), Ministry of Education, Wuhan, Hubei, China
| | - Linhui Wu
- Institute of Stem Cell and Regenerative Biology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction (Huazhong Agricultural University), Ministry of Education, Wuhan, Hubei, China
| | - Kai Zhou
- Institute of Stem Cell and Regenerative Biology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction (Huazhong Agricultural University), Ministry of Education, Wuhan, Hubei, China
| | - Zhengang Fan
- Institute of Stem Cell and Regenerative Biology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction (Huazhong Agricultural University), Ministry of Education, Wuhan, Hubei, China
| | - Tingting Wang
- Institute of Stem Cell and Regenerative Biology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction (Huazhong Agricultural University), Ministry of Education, Wuhan, Hubei, China
| | - Taotao Hu
- Institute of Stem Cell and Regenerative Biology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction (Huazhong Agricultural University), Ministry of Education, Wuhan, Hubei, China
| | - Xia Zhang
- Institute of Stem Cell and Regenerative Biology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction (Huazhong Agricultural University), Ministry of Education, Wuhan, Hubei, China
| | - Jilong Zhou
- Institute of Stem Cell and Regenerative Biology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction (Huazhong Agricultural University), Ministry of Education, Wuhan, Hubei, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Yi-Liang Miao
- Institute of Stem Cell and Regenerative Biology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction (Huazhong Agricultural University), Ministry of Education, Wuhan, Hubei, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Huazhong Agricultural University, Wuhan, Hubei, China
| |
Collapse
|
17
|
Akhtar MJ, Khan SA, Kumar B, Chawla P, Bhatia R, Singh K. Role of sodium dependent SLC13 transporter inhibitors in various metabolic disorders. Mol Cell Biochem 2022:10.1007/s11010-022-04618-7. [DOI: 10.1007/s11010-022-04618-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 11/23/2022] [Indexed: 12/14/2022]
|
18
|
A Novel and Cross-Species Active Mammalian INDY (NaCT) Inhibitor Ameliorates Hepatic Steatosis in Mice with Diet-Induced Obesity. Metabolites 2022; 12:metabo12080732. [PMID: 36005604 PMCID: PMC9413491 DOI: 10.3390/metabo12080732] [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: 06/13/2022] [Revised: 07/31/2022] [Accepted: 08/02/2022] [Indexed: 11/25/2022] Open
Abstract
Mammalian INDY (mINDY, NaCT, gene symbol SLC13A5) is a potential target for the treatment of metabolically associated fatty liver disease (MAFLD). This study evaluated the effects of a selective, cross-species active, non-competitive, non-substrate-like inhibitor of NaCT. First, the small molecule inhibitor ETG-5773 was evaluated for citrate and succinate uptake and fatty acid synthesis in cell lines expressing both human NaCT and mouse Nact. Once its suitability was established, the inhibitor was evaluated in a diet-induced obesity (DIO) mouse model. DIO mice treated with 15 mg/kg compound ETG-5773 twice daily for 28 days had reduced body weight, fasting blood glucose, and insulin, and improved glucose tolerance. Liver triglycerides were significantly reduced, and body composition was improved by reducing fat mass, supported by a significant reduction in the expression of genes for lipogenesis such as SREBF1 and SCD1. Most of these effects were also evident after a seven-day treatment with the same dose. Further mechanistic investigation in the seven-day study showed increased plasma β-hydroxybutyrate and activated hepatic adenosine monophosphate-activated protein kinase (AMPK), reflecting findings from Indy (−/−) knockout mice. These results suggest that the inhibitor ETG-5773 blocked citrate uptake mediated by mouse and human NaCT to reduce liver steatosis and body fat and improve glucose regulation, proving the concept of NaCT inhibition as a future liver treatment for MAFLD.
Collapse
|
19
|
Lactucin & Lactucopicrin ameliorates FFA-induced steatosis in HepG2 cells via modulating lipid metabolism. J Pharmacol Sci 2022; 150:110-122. [DOI: 10.1016/j.jphs.2022.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 07/09/2022] [Accepted: 07/28/2022] [Indexed: 11/19/2022] Open
|
20
|
Batchuluun B, Pinkosky SL, Steinberg GR. Lipogenesis inhibitors: therapeutic opportunities and challenges. Nat Rev Drug Discov 2022; 21:283-305. [PMID: 35031766 PMCID: PMC8758994 DOI: 10.1038/s41573-021-00367-2] [Citation(s) in RCA: 206] [Impact Index Per Article: 68.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2021] [Indexed: 12/12/2022]
Abstract
Fatty acids are essential for survival, acting as bioenergetic substrates, structural components and signalling molecules. Given their vital role, cells have evolved mechanisms to generate fatty acids from alternative carbon sources, through a process known as de novo lipogenesis (DNL). Despite the importance of DNL, aberrant upregulation is associated with a wide variety of pathologies. Inhibiting core enzymes of DNL, including citrate/isocitrate carrier (CIC), ATP-citrate lyase (ACLY), acetyl-CoA carboxylase (ACC) and fatty acid synthase (FAS), represents an attractive therapeutic strategy. Despite challenges related to efficacy, selectivity and safety, several new classes of synthetic DNL inhibitors have entered clinical-stage development and may become the foundation for a new class of therapeutics. De novo lipogenesis (DNL) is vital for the maintenance of whole-body and cellular homeostasis, but aberrant upregulation of the pathway is associated with a broad range of conditions, including cardiovascular disease, metabolic disorders and cancers. Here, Steinberg and colleagues provide an overview of the physiological and pathological roles of the core DNL enzymes and assess strategies and agents currently in development to therapeutically target them.
Collapse
Affiliation(s)
- Battsetseg Batchuluun
- Centre for Metabolism, Obesity and Diabetes Research, Department of Medicine and Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | | | - Gregory R Steinberg
- Centre for Metabolism, Obesity and Diabetes Research, Department of Medicine and Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
21
|
Zhan Z, Li A, Zhang W, Wu X, He J, Li Z, Li Y, Sun J, Zhang H. ATP-citrate lyase inhibitor improves ectopic lipid accumulation in the kidney in a db/db mouse model. Front Endocrinol (Lausanne) 2022; 13:914865. [PMID: 36568100 PMCID: PMC9771989 DOI: 10.3389/fendo.2022.914865] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 11/11/2022] [Indexed: 12/12/2022] Open
Abstract
AIM We evaluated a novel treatment for obesity-related renal, an ATP-citrate lyase (ACL) inhibitor, to attenuate ectopic lipid accumulation (ELA) in the kidney and the ensuing inflammation. MATERIALS AND METHODS An ACL inhibitor was administered intragastrically to 12-week-old db/db mice for 30 days. The appearance of ELA was observed by staining kidney sections with Oil Red O, and the differences in tissue lipid metabolites were assessed by mass spectrometry. The anti-obesity and renoprotection effects of ACL inhibitors were observed by histological examination and multiple biochemical assays. RESULTS Using the AutoDock Vina application, we determined that among the four known ACL inhibitors (SB-204990, ETC-1002, NDI-091143, and BMS-303141), BMS-303141 had the highest affinity for ACL and reduced ACL expression in the kidneys of db/db mice. We reported that BMS-303141 administration could decrease the levels of serum lipid and renal lipogenic enzymes acetyl-CoA carboxylase (ACC), fatty acid synthase (FAS), HMG-CoA reductase (HMGCR), and diminish renal ELA in db/db mice. In addition, we found that reducing ELA improved renal injuries, inflammation, and tubulointerstitial fibrosis. CONCLUSION ACL inhibitor BMS-303141 protects against obesity-related renal injuries.
Collapse
Affiliation(s)
- Zishun Zhan
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- The Critical Kidney Disease Research Center of Central South University, Changsha, Hunan, China
| | - Aimei Li
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- The Critical Kidney Disease Research Center of Central South University, Changsha, Hunan, China
| | - Wei Zhang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- The Critical Kidney Disease Research Center of Central South University, Changsha, Hunan, China
| | - Xueqin Wu
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- The Critical Kidney Disease Research Center of Central South University, Changsha, Hunan, China
| | - Jinrong He
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- The Critical Kidney Disease Research Center of Central South University, Changsha, Hunan, China
| | - Zhi Li
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- The Critical Kidney Disease Research Center of Central South University, Changsha, Hunan, China
| | - Yanchun Li
- Division of Biological Sciences, Department of Medicine, University of Chicago, Chicago, Chicago, IL, United States
| | - Jian Sun
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- The Critical Kidney Disease Research Center of Central South University, Changsha, Hunan, China
- Department of Rheumatology and Immunology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- *Correspondence: Hao Zhang, ; Jian Sun,
| | - Hao Zhang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- The Critical Kidney Disease Research Center of Central South University, Changsha, Hunan, China
- *Correspondence: Hao Zhang, ; Jian Sun,
| |
Collapse
|