1
|
Ma Y, Du C, Liu Y, Feng M, Shou Y, Yu D, Jin Y. Aristolochic acid-induced dyslipidemia and hepatotoxicity: The potential role of FXR and AHR receptors. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 287:117266. [PMID: 39509784 DOI: 10.1016/j.ecoenv.2024.117266] [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: 07/22/2024] [Revised: 10/26/2024] [Accepted: 10/27/2024] [Indexed: 11/15/2024]
Abstract
Aristolochic acids (AAs) represent a class of nitrophenanthrene carboxylic acids naturally existing or accidentally mixed in herbal medicines or crops, which have long been recognized for causing nephropathy. Recently, the linkage between AAs and liver injury has become a concern; however, the current understanding of the mechanism or mode of action (MOA) is limited. In the present study, we investigated nuclear receptor-mediated MOA associated with AAs-induced liver injury including dyslipidemia and hepatotoxicity. Bioinformatic analysis of AAI-interacting genes indicated nuclear receptor-mediated metabolizing pathways; Transcriptomic profiling of AAs-exposed rats with liver injury suggested FXR-, NRF2-, and AHR- mediated pathways in the injured livers of the rats. Mechanistic investigation using HepG2 cells indicated AAI-induced hepatic lipid accumulation by elevating Triglyceride (TG) through inhibition of the FXR. In addition, AAI-induced hepatocellular damage by activating the AHR pathway, which further generated ROS and activated the NRF2 pathway. Together, these results provided new clues for researchers who are interested in chemical-induced liver injury.
Collapse
Affiliation(s)
- Yumei Ma
- School of Public Health, Qingdao University, Qingdao, China
| | - Chenlong Du
- Ningxia Hui Autonomous Region Center for Disease Control and Prevention, Yinchuan, China
| | - Yuzhen Liu
- Gaomi Municipal Center for Disease Control and Prevention, Weifang Institute of Preventive Medicine, Weifang, China
| | - Meiyao Feng
- Department of Environmental Health, Qingdao Municipal Center for Disease Control and Prevention, Qingdao Institute of Preventive Medicine, Qingdao, China
| | - Yingqing Shou
- School of Public Health, Qingdao University, Qingdao, China
| | - Dianke Yu
- School of Public Health, Qingdao University, Qingdao, China
| | - Yuan Jin
- School of Public Health, Qingdao University, Qingdao, China.
| |
Collapse
|
2
|
Udayan SP, Hariharan S, Nevin KG. Multifaceted bioactivity of marine fungal derived secondary metabolite, xyloketal B -a review. Toxicol Res (Camb) 2024; 13:tfae156. [PMID: 39345795 PMCID: PMC11425363 DOI: 10.1093/toxres/tfae156] [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: 04/01/2024] [Revised: 09/03/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
Background A growing number of findings have focused on the distinctive physiochemical characteristics that marine microorganisms have acquired as a result of their adaptation to the challenging conditions inherent in the marine environment. It has been established that the marine environment is a very rich source of bioactive substances with a variety of biological effects and structural diversity. A major discovery was the extraction of xyloketals from Xylaria sp. Numerous thorough studies have subsequently been carried out to determine the medicinal potential of these bioactive components. Xyloketals are thought to be a very promising and significant class of naturally occurring substances with a wide range of potent biological activities, such as radical scavenging, suppression of cell proliferation, reduction of neonatal hypoxic-ischemic brain injury, antioxidant activity, inhibition of acetylcholine esterase, inhibition of L-calcium channels, and others. Xyloketal B is one of the most potent molecules with significant therapeutic properties among the numerous variants discovered. Conclusion This review summarizes the structural characterization of all naturally occurring xyloketal compounds, especially the B derivative with an emphasis on their bioactivity and provides an outline of how xyloketals operate in diverse disease scenarios.
Collapse
Affiliation(s)
- Sreelekshmi Puthuvalnikarthil Udayan
- Department of Marine Biosciences, Faculty of Ocean Science and Technology, Kerala University of Fisheries and Ocean Studies, Panangad PO, Kochi, Ernakulam District, Kerala 682506, India
| | - Sini Hariharan
- Department of Biochemistry, Government College, Kariavattom PO, Thiruvananthapuram District, Kerala 695581, India
| | - Kottayath Govindan Nevin
- Department of Marine Biosciences, Faculty of Ocean Science and Technology, Kerala University of Fisheries and Ocean Studies, Panangad PO, Kochi, Ernakulam District, Kerala 682506, India
- Centre for Bioactive Substances from Marine Organisms, Kerala University of Fisheries and Ocean Studies, Kochi 682506, India
| |
Collapse
|
3
|
Yan M, Zhao Y, Man S, Dai Y, Ma L, Gao W. Diosgenin as a substitute for cholesterol alleviates NAFLD by affecting CYP7A1 and NPC1L1-related pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 125:155299. [PMID: 38301301 DOI: 10.1016/j.phymed.2023.155299] [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: 04/14/2023] [Revised: 12/08/2023] [Accepted: 12/17/2023] [Indexed: 02/03/2024]
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) rapidly becomes the leading cause of end-stage liver disease or liver transplantation. Nowadays, there has no approved drug for NAFLD treatment. Diosgenin as the structural analogue of cholesterol attenuates hypercholesterolemia by inhibiting cholesterol metabolism, which is an important pathogenesis in NAFLD progression. However, there has been no few report concerning its effects on NAFLD so far. METHODS Using a high-fat diet & 10% fructose-feeding mice, we evaluated the anti-NAFLD effects of diosgenin. Transcriptome sequencing, LC/MS analysis, molecular docking simulation, molecular dynamics simulations and Luci fluorescent reporter gene analysis were used to evaluate pathways related to cholesterol metabolism. RESULTS Diosgenin treatment ameliorated hepatic dysfunction and inhibited NAFLD formation including lipid accumulation, inflammation aggregation and fibrosis formation through regulating cholesterol metabolism. For the first time, diosgenin was structurally similar to cholesterol, down-regulated expression of CYP7A1 and regulated cholesterol metabolism in the liver (p < 0.01) and further affecting bile acids like CDCA, CA and TCA in the liver and feces. Besides, diosgenin decreased expression of NPC1L1 and suppressed cholesterol transport (p < 0.05). Molecular docking and molecular dynamics further proved that diosgenin was more strongly bound to CYP7A1. Luci fluorescent reporter gene analysis revealed that diosgenin concentration-dependently inhibited the enzymes activity of CYP7A1. CONCLUSION Our findings demonstrated that diosgenin was identified as a specific regulator of cholesterol metabolism, which pave way for the design of novel clinical therapeutic strategies.
Collapse
Affiliation(s)
- Mengyao Yan
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China.
| | - Yixin Zhao
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Shuli Man
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China.
| | - Yujie Dai
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Long Ma
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Wenyuan Gao
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, Tianjin 300072, China
| |
Collapse
|
4
|
Dai X, Li X, Yin D, Chen X, Wang L, Pang L, Fu Y. Identification and characterization of TOR in Macrobrachium rosenbergii and its role in muscle protein and lipid production. Sci Rep 2024; 14:2082. [PMID: 38267514 PMCID: PMC10810085 DOI: 10.1038/s41598-023-50300-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 12/18/2023] [Indexed: 01/26/2024] Open
Abstract
The recent scarcity of fishmeal and other resources means that studies on the intrinsic mechanisms of nutrients in the growth and development of aquatic animals at the molecular level have received widespread attention. The target of rapamycin (TOR) pathway has been reported to receive signals from nutrients and environmental stresses, and regulates cellular anabolism and catabolism to achieve precise regulation of cell growth and physiological activities. In this study, we cloned and characterized the full-length cDNA sequence of the TOR gene of Macrobrachium rosenbergii (MrTOR). MrTOR was expressed in all tissues, with higher expression in heart and muscle tissues. In situ hybridization also indicated that MrTOR was expressed in muscle, mainly around the nucleus. RNA interference decreased the expression levels of MrTOR and downstream protein synthesis-related genes (S6K, eIF4E, and eIF4B) (P < 0.05) and the expression and enzyme activity of the lipid synthesis-related enzyme, fatty acid synthase (FAS), and increased enzyme activity of the lipolysis-related enzyme, lipase (LPS). In addition, amino acid injection significantly increased the transcript levels of MrTOR and downstream related genes (S6K, eIF4E, eIF4B, and FAS), as well as triglyceride and total cholesterol tissue levels and FAS activity. Starvation significantly increased transcript levels and enzyme activities of adenylate-activated protein kinase and LPS and decreased transcript levels and enzyme activities of FAS, as well as transcript levels of MrTOR and its downstream genes (P < 0.05), whereas amino acid injection alleviated the starvation-induced decreases in transcript levels of these genes. These results suggested that arginine and leucine activated the TOR signaling pathway, promoted protein and lipid syntheses, and alleviated the pathway changes induced by starvation.
Collapse
Affiliation(s)
- Xilin Dai
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, 201306, China.
- Shanghai Collaborative Innovation for Aquatic Animal Genetics and Breeding, Shanghai, 201306, China.
- National Experimental Teaching Demonstration Centre for Aquatic Sciences, Shanghai Ocean University, Shanghai, 201306, China.
| | - Xuenan Li
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, 201306, China
- Shanghai Collaborative Innovation for Aquatic Animal Genetics and Breeding, Shanghai, 201306, China
| | - Danhui Yin
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, 201306, China
- Shanghai Collaborative Innovation for Aquatic Animal Genetics and Breeding, Shanghai, 201306, China
| | - Xin Chen
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, 201306, China
- Shanghai Collaborative Innovation for Aquatic Animal Genetics and Breeding, Shanghai, 201306, China
| | - Linwei Wang
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, 201306, China
- Shanghai Collaborative Innovation for Aquatic Animal Genetics and Breeding, Shanghai, 201306, China
| | - Luyao Pang
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, 201306, China
- Shanghai Collaborative Innovation for Aquatic Animal Genetics and Breeding, Shanghai, 201306, China
| | - Yuanshuai Fu
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, 201306, China
- Shanghai Collaborative Innovation for Aquatic Animal Genetics and Breeding, Shanghai, 201306, China
- National Experimental Teaching Demonstration Centre for Aquatic Sciences, Shanghai Ocean University, Shanghai, 201306, China
| |
Collapse
|
5
|
Dai Y, Zhang X, Xu Y, Wu Y, Yang L. The Protective Effects of Cinnamyl Alcohol Against Hepatic Steatosis, Oxidative and Inflammatory Stress in Nonalcoholic Fatty Liver Disease Induced by Childhood Obesity. Immunol Invest 2023; 52:1008-1022. [PMID: 37962037 DOI: 10.1080/08820139.2023.2280248] [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: 11/15/2023]
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is characterized by excessive intracellular lipid accumulation, oxidative stress, and inflammation. Cinnamyl alcohol (CA), one of the cinnamon extracts, has been shown to exhibit anti-oxidative and anti-inflammatory activities. We proposed that CA was beneficial to NAFLD. METHODS Serum cytokines and components of the lipid metabolism were determined in children with NAFLD against age-matched comparisons. A NAFLD mouse model was established by high fat and high carbohydrate (HFHC) diet in male C57BL/6 mouse pups, followed by administration of CA. The effects of CA on lipid metabolism, oxidative stress, and inflammation in hepatic tissues were assessed. RESULTS Abnormal lipid metabolism and inflammatory responses were observed in the children with NAFLD as compared with the controls. CA reduced the weight of obese mice without affecting food intake as well as alleviating liver injury caused by HFHC feeding. CA was found to mitigate dyslipidemia and reduce hepatic steatosis in HFHC-fed mice by down-regulating genes related to lipogenesis, including peroxisome proliferator-activated receptor gamma (PPARγ), sterol regulatory element-binding transcription factor-1c (SREBP-1c), and acetyl-CoA carboxylase 1 (ACC1). Additionally, CA treatment reversed HFHC-induced oxidative stress and inflammation, evidenced by the decreased liver reactive oxygen species (ROS), hepatic inflammatory cytokine levels, and F4/80-positive macrophage infiltration in HFHC diet mice. CA reduced the protein levels of pyrin domain-containing protein 3 (NLRP3), adapter protein apoptosis-associated speck-like protein (ASC), and caspase-1 in the liver tissues significantly. CONCLUSION CA alleviates HFHC-induced NAFLD in mice, which is associated with the amelioration in lipid metabolism, oxidative stress, and inflammation.
Collapse
Affiliation(s)
- Yu Dai
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Pediatrics, Anhui Public Health Clinical Center, Hefei, China
| | - Xuemin Zhang
- Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Pediatrics, Anhui Public Health Clinical Center, Hefei, China
| | - Yao Xu
- Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Pediatrics, Anhui Public Health Clinical Center, Hefei, China
| | - Ya Wu
- Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Pediatrics, Anhui Public Health Clinical Center, Hefei, China
| | - Liqi Yang
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
6
|
Zhang M, Zou X, Du Y, Pan Z, He F, Sun Y, Li M. Integrated Transcriptomics and Metabolomics Reveal the Mechanism of Alliin in Improving Hyperlipidemia. Foods 2023; 12:3407. [PMID: 37761116 PMCID: PMC10528980 DOI: 10.3390/foods12183407] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
This research aims to assess the anti-hyperlipidemia effects of alliin in vivo and its potential mechanisms through transcriptomics and metabolomics analysis. A hyperlipidemia mode was established in C57BL/6 mice fed a high-fat diet, and the related physiological parameters of the animals were recorded. Serum TC and MDA in livers significantly decreased by 12.34% and 29.59%, respectively, and SOD and CAT in livers significantly increased by 40.64% and 39.05%, respectively, after high doses of alliin interventions. In total, 148 significantly different genes, particularly Cel, Sqle, Myc, and Ugt1a2, were revealed for their potential roles in HFD-induced alliin, mainly through steroid biosynthesis, triglyceride metabolism, drug metabolism-cytochrome P450, and the PI3K-Akt signaling pathway, according to transcriptomics analysis. Metabolomics results revealed 18 significantly different metabolites between the alliin group and HFD group, which were classified as carboxylic acids, such as N-undecanoylglycine, adipic acid, D-pantothenic acid, cyprodenate, and pivagabine. We found pantothenic acid played a vital role and was effective through pantothenic acid and CoA biosynthesis metabolism. The "steroid biosynthesis pathway" was identified as the most significant metabolic pathway by integrated transcriptomics and metabolomics analysis. This work offered a theoretical framework for the mechanism of alliin lipid lowering in the future. The development and utilization of alliin will be a viable strategy to improve the health status of people with hyperlipidemia, suggesting prospective market opportunities.
Collapse
Affiliation(s)
- Min Zhang
- Guangdong Provincial Key Lab of Food Safety and Quality, South China Agricultural University, Guangzhou 510642, China; (M.Z.); (X.Z.); (Y.D.); (Z.P.); (Y.S.)
- College of Food, South China Agricultural University, Guangzhou 510642, China
| | - Xiaoying Zou
- Guangdong Provincial Key Lab of Food Safety and Quality, South China Agricultural University, Guangzhou 510642, China; (M.Z.); (X.Z.); (Y.D.); (Z.P.); (Y.S.)
- College of Food, South China Agricultural University, Guangzhou 510642, China
| | - Yixuan Du
- Guangdong Provincial Key Lab of Food Safety and Quality, South China Agricultural University, Guangzhou 510642, China; (M.Z.); (X.Z.); (Y.D.); (Z.P.); (Y.S.)
- College of Food, South China Agricultural University, Guangzhou 510642, China
| | - Zhuangguang Pan
- Guangdong Provincial Key Lab of Food Safety and Quality, South China Agricultural University, Guangzhou 510642, China; (M.Z.); (X.Z.); (Y.D.); (Z.P.); (Y.S.)
- College of Food, South China Agricultural University, Guangzhou 510642, China
| | - Fangqing He
- Guangdong Provincial Key Lab of Food Safety and Quality, South China Agricultural University, Guangzhou 510642, China; (M.Z.); (X.Z.); (Y.D.); (Z.P.); (Y.S.)
- College of Food, South China Agricultural University, Guangzhou 510642, China
| | - Yuanming Sun
- Guangdong Provincial Key Lab of Food Safety and Quality, South China Agricultural University, Guangzhou 510642, China; (M.Z.); (X.Z.); (Y.D.); (Z.P.); (Y.S.)
- College of Food, South China Agricultural University, Guangzhou 510642, China
| | - Meiying Li
- Guangdong Provincial Key Lab of Food Safety and Quality, South China Agricultural University, Guangzhou 510642, China; (M.Z.); (X.Z.); (Y.D.); (Z.P.); (Y.S.)
- College of Food, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
7
|
Romero EO, Perkins JC, Burch JE, Delgadillo DA, Nelson HM, Narayan ARH. Chemoenzymatic Synthesis of (+)-Xyloketal B. Org Lett 2023; 25:1547-1552. [PMID: 36827601 DOI: 10.1021/acs.orglett.3c00334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2023]
Abstract
Xyloketal B is a pentacyclic fungal marine natural product that has shown potential for the treatment of diseases such as Alzheimer's disease and atherosclerosis. Herein, we describe the first asymmetric synthesis of this natural product, which relies on a chemoenzymatic strategy. This approach leverages a biocatalytic benzylic hydroxylation to access to an ortho-quinone methide intermediate which is captured in a [4 + 2] cycloaddition to stereoselectively yield a key cyclic ketal intermediate enroute to (+)-xyloketal B. The relative configuration of this intermediate was rapidly confirmed as the desired stereoisomer using MicroED. To complete the synthesis, a second ortho-quinone methide was accessed through a reductive approach, ultimately leading to the stereoselective synthesis of (+)-xyloketal B.
Collapse
Affiliation(s)
- Evan O Romero
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States.,Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jonathan C Perkins
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States.,Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jessica E Burch
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California 91125, United States
| | - David A Delgadillo
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California 91125, United States
| | - Hosea M Nelson
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California 91125, United States
| | - Alison R H Narayan
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States.,Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, United States.,Program in Chemical Biology, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
8
|
Ning DS, Chen YJ, Lin CJ, Wang CC, Zhao HW, Wang KT, Lee MC, Tayo LL, Chiu WC, Yeh CL, Lee CJ. Hepatoprotective effect of botanical drug formula on high-fat diet-induced non-alcoholic fatty liver disease by inhibiting lipogenesis and promoting anti-oxidation. Front Pharmacol 2022; 13:1026912. [PMID: 36506588 PMCID: PMC9729544 DOI: 10.3389/fphar.2022.1026912] [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: 08/24/2022] [Accepted: 10/31/2022] [Indexed: 11/27/2022] Open
Abstract
With the prevalence of obesity and other components of metabolic syndrome, Non-alcoholic fatty liver disease (NAFLD) has become increasingly common. In recent years, much attention has been paid to various plant sources, hoping to find a treatment for NAFLD in plants. The Livsooth authentic herbal formula (LAH, ), a botanical drug formula combined with Puerariae lobatae radix, Lonicerae japonicae flos, Hoveniae semen, and Siraitiae fructus. This study used a network pharmacology approach to predict the potential mechanisms of LAH against NAFLD. Gene Ontology (GO) and KEGG pathway enrichment analyses have identified potential biochemical and signaling pathways. Subsequently, the potential mechanism of action of LAH on NAFLD predicted by network pharmacology analysis was validated in a high-fat diet (HFD)-induced NAFLD model in C57BL/6 mice. Our results demonstrated that LAH ameliorated hepatocyte steatosis in liver tissue by activating the AMPK pathway and decreasing serum triglycerides, low-density lipoprotein, glucose, and cholesterol. Besides, LAH increased the hepatic antioxidant enzymes activities, suggested that LAH improved oxidative stress markers in HFD induced NAFLD mice. In vitro experiments confirmed that the active component of LAH, puerarin, regulates lipid accumulation through the AMPK pathway. In conclusion, our study shows that network pharmacology predictions are consistent with experimental validation. LAH can be a candidate supplement for the prevention of NAFLD.
Collapse
Affiliation(s)
- De-Shan Ning
- Infinitus (China) Company Ltd., Guangzhou, China
| | - Yu-Ju Chen
- Ph.D. Program in Clinical Drug Development of Herbal Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chien-Ju Lin
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ching-Chiung Wang
- Ph.D. Program in Clinical Drug Development of Herbal Medicine, Taipei Medical University, Taipei, Taiwan,Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, Taiwan,School of Pharmacy, Taipei Medical University, Taipei, Taiwan,Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | | | | | | | - Lemmuel L. Tayo
- School of Chemical, Biological Materials Science and Engineering, Mapúa University, Manila, Philippines
| | - Wan-Chun Chiu
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei, Taiwan,Department of Nutrition, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Chiu-Li Yeh
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei, Taiwan
| | - Chia-Jung Lee
- Ph.D. Program in Clinical Drug Development of Herbal Medicine, Taipei Medical University, Taipei, Taiwan,Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, Taiwan,Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei, Taiwan,*Correspondence: Chia-Jung Lee,
| |
Collapse
|
9
|
Tang Z, Li L, Xia Z. Exploring Anti-Nonalcoholic Fatty Liver Disease Mechanism of Gardeniae Fructus by Network Pharmacology, Molecular Docking, and Experiment Validation. ACS OMEGA 2022; 7:25521-25531. [PMID: 35910181 PMCID: PMC9330257 DOI: 10.1021/acsomega.2c02629] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 07/01/2022] [Indexed: 05/08/2023]
Abstract
Gardeniae fructus (GF), the fruit from Gardenia jasminoides Ellis, is a traditional Chinese medicine used for the treatment of nonalcoholic fatty liver disease (NAFLD) in the clinic. To explore the hepatoprotective mechanism of GF for the treatment of NAFLD, we proposed a novel strategy that integrated in vivo efficacy evaluation, network pharmacology analysis, molecular docking, and experimental validation. A NAFLD animal model induced by high fat diet (HFD) feed was established, then orally administrated with or without GF. The results showed that GF significantly decreased the levels of serum total cholesterol (TC), lipoprotein cholesterol, triglyceride (TG), alanine aminotransferase, aspartate aminotransferase, lactate dehydrogenase, free fatty acids, glucose, and insulin and the levels of liver TG, TC, and malondialdehyde compared with the nontreated HFD group. Network pharmacology studies showed that quercetin, oleanolic acid, kaempferol, and geniposide were the main biocompounds in GF that targeted the PPARα and PPARγ genes through regulating the PPAR and AMPK signal pathways to protect against NAFLD. The interactions between bioactive compounds and their corresponding target proteins were analyzed by molecular docking and subsequently confirmed using the qRT-PCR assay. Collectively, GF was a therapeutic drug for the treatment of NAFLD.
Collapse
Affiliation(s)
- Zhongyan Tang
- Department
of Emergency and Critical Care Medicine, Jin Shan Hospital, Fudan University, Shanghai 201508, China
| | - Lin Li
- Department
of Operative Dentistry and Endodontics, School and Hosipital of Stomatology,
Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Tongji University, 399 Middle Yan Chang Road, Shanghai 200072, China
| | - Zhengxiang Xia
- Department
of Pharmacy, School and Hosipital of Stomatology, Shanghai Engineering
Research Center of Tooth Restoration and Regeneration, Tongji University, 399 Middle Yan Chang Road, Shanghai 200072, China
- . Tel: +8621-66315500
| |
Collapse
|
10
|
Tong Y, Zhu W, Wen T, Mukhamejanova Z, Xu F, Xiang Q, Pang J. Xyloketal B Reverses Nutritional Hepatic Steatosis, Steatohepatitis, and Liver Fibrosis through Activation of the PPARα/PGC1α Signaling Pathway. JOURNAL OF NATURAL PRODUCTS 2022; 85:1738-1750. [PMID: 35749236 DOI: 10.1021/acs.jnatprod.2c00259] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) represents a class of disorders including hepatic steatosis, steatohepatitis, and liver fibrosis. Previous research suggested that xyloketal B (Xyl-B), a marine-derived natural product, could attenuate the NAFLD-related lipid accumulation. Herein, we investigated the protective mechanism of Xyl-B in a high-fat diet (HFD) mice fatty liver model by combining a quantitative proteomic approach with experimental methods. The results showed that the administration of Xyl-B (20 and 40 mg·kg-1·day-1, ip) ameliorated the hepatic steatosis in HFD mice. Proteomic profiling together with bioinformatics analysis highlighted the upregulation of a cluster of peroxisome proliferator-activated receptor-α (PPARα) downstream enzymes mainly related to fatty acid oxidation (FAO) as key changes after the treatment. These changes were subsequently confirmed by bioassays. Moreover, further results showed that the expression levels of PPARα and PPARγ coactivator-1α (PGC1α) were increased after the treatment. The related mode-of-action was confirmed by PPARα inhibition. Furthermore, we evaluated the PPARα-mediated anti-inflammatory and antifibrosis effect of Xyl-B in methionine-choline-deficient (MCD) mice hepatitis and liver fibrosis models. According to the results, the histological features were improved, and the levels of inflammatory factors, adhesion molecules, as well as fibrosis markers were decreased after the treatment. Collectively, these results indicated that Xyl-B ameliorated different phases of NAFLD through activation of the PPARα/PGC1α signaling pathway. Our findings revealed the possible metabolism-regulating mechanism of Xyl-B, broadened the application of xyloketal family compounds, and may provide a new strategy to curb the development of NAFLD.
Collapse
Affiliation(s)
- Yichen Tong
- School of Chemistry, Sun Yat-sen University, Guangzhou 510006, China
| | - Wentao Zhu
- School of Chemistry, Sun Yat-sen University, Guangzhou 510006, China
| | - Tianzhi Wen
- School of Chemistry, Sun Yat-sen University, Guangzhou 510006, China
| | | | - Fang Xu
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE) & Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Qi Xiang
- Institute of Biomedicine & Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Jiyan Pang
- School of Chemistry, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
11
|
Yan X, Zhang Y, Peng Y, Li X. The water extract of Radix scutellariae, its total flavonoids and baicalin inhibited CYP7A1 expression, improved bile acid, and glycolipid metabolism in T2DM mice. JOURNAL OF ETHNOPHARMACOLOGY 2022; 293:115238. [PMID: 35351576 DOI: 10.1016/j.jep.2022.115238] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 03/11/2022] [Accepted: 03/23/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Radix scutellariae (the root of Scutellaria baicalensis Georgi), is a traditional Chinese medicine (TCM) used to treat type 2 diabetes mellitus (T2DM). Abundant flavonoids are the antidiabetic components of Radix scutellariae, of which baicalin (Baicalein 7-O-glucuronide, BG) is the major bioactive component. Our previous studies found that the water extract of Radix scutellariae (WESB) could exert hypoglycemic and hypolipidemic efficacies by adjusting the ileum FXR-medicated interaction between gut microbiota and bile acid (BA) metabolism. However, it remains unclear whether WESB and its biologically active ingredients exert an antidiabetic effect through bile acid signaling mediated by FXR-CYP7A1. AIMS OF THE STUDY To explore the mechanism of WESB and its total flavonoids (TF) further and BG on BA signals and glycolipid metabolism in T2DM mice. MATERIALS AND METHODS The antidiabetic effects of WESB, TF and BG were evaluated by indexing the body weight, fasting blood glucose (FBG) and oral glucose tolerance test (OGTT) in HFD/STZ-induced (high-fat diet and streptozocin) diabetic mice, and comparing them with the positive control (metformin). The lipids in the mouse liver and the total bile acids (TBA) in the mouse liver and bile were detected by commercial kits. The concentration of BAs in the mouse feces was determined by liquid chromatography-tandem mass spectrometry. The protein expression levels of cholesterol 7α-hydroxylase (CYP7A1), farnesol X receptor (FXR), etc., in the liver and/or ileum, play a key role in the BAs metabolism of T2DM mice were evaluated by immunoblot analysis. RESULTS The hyperglycemia and impaired glucose tolerance of T2DM mice were improved after WESB, TF and BG treatment. Especially after BG administration, the levels of low-density lipoprotein-cholesterol (LDL-c) and total glyceride (TG) in the T2DM mouse liver were significantly decreased (p < 0.05). While the level of high-density lipoprotein cholesterol (HDL-c) was significant increased (p < 0.001). Meanwhile, the levels of TBA in both the liver and bile of T2DM mice were significantly decreased by BG (p < 0.05). Moreover, the high expression of CYP7A1 in the liver of T2DM mice was significantly inhibited by WESB, TF and BG (p < 0.05), and the high expression of FXR in the ileum of T2DM mice was significantly inhibited by TF (p < 0.05). CONCLUSION These results indicated that the hypoglycemic effects of WESB, TF and BG might be exerted by inhibiting the expression of CYP7A1 in T2DM mice, and TF inhibited expression of intestinal FXR by inducing changes in fecal BA profile. BG significantly improved hepatic lipid metabolism. Moreover, BG reduced lipid accumulation in the liver and bile by inhibiting the expression of CYP7A1 in T2DM mice. These findings provide useful explanations for the antidiabetic mechanism of Radix scutellariae.
Collapse
Affiliation(s)
- Xiumei Yan
- School of Pharmacy, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Minhang District, Shanghai, 200240, China
| | - Yulong Zhang
- School of Pharmacy, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Minhang District, Shanghai, 200240, China
| | - Ying Peng
- School of Pharmacy, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Minhang District, Shanghai, 200240, China
| | - Xiaobo Li
- School of Pharmacy, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Minhang District, Shanghai, 200240, China.
| |
Collapse
|
12
|
Xu Z, Hu W, Wang B, Xu T, Wang J, Wei D. Canagliflozin Ameliorates Nonalcoholic Fatty Liver Disease by Regulating Lipid Metabolism and Inhibiting Inflammation through Induction of Autophagy. Yonsei Med J 2022; 63:619-631. [PMID: 35748073 PMCID: PMC9226837 DOI: 10.3349/ymj.2022.63.7.619] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 01/17/2022] [Accepted: 02/09/2022] [Indexed: 12/28/2022] Open
Abstract
PURPOSE Nonalcoholic fatty liver disease (NAFLD) is closely associated with metabolic diseases, including obesity and diabetes, and has gradually become the most common cause of chronic liver disease. We investigated the effects of sodium glucose cotransporter 2 (SGLT2) inhibitor canagliflozin on NAFLD in high-fat diet (HFD)-induced obese mice and possible underlying mechanisms. MATERIALS AND METHODS Male C57BL/6 mice were fed a normal-diet, HFD, or HFD with canagliflozin for 14 weeks. AML-12 hepatocytes were treated with canagliflozin. Expression of related pathways was assessed. RESULTS Canagliflozin administration reduced body weight and fat mass, compared with HFD alone. Canagliflozin improved glucose and lipid metabolic disorders. Compared with HFD-fed mice, liver weight, serum alanine transaminase (ALT) levels, and hepatic lipid accumulation were decreased after canagliflozin administration. Additionally, canagliflozin upregulated lipolysis markers (CPT1a, ACOX1, and ACADM), downregulated lipogenesis markers (SREBP-1c and FASN), and suppressed the production of inflammatory cytokines (TNFα, MCP1, IL-1β, and IL-6), consistent with significantly increased LC3 II/I and Atg7 levels in the liver following canagliflozin treatment. In vitro, canagliflozin increased CPT1a, ACOX1, and ACADM expression, decreased SREBP-1c and FASN protein expression, and reduced TNFα, MCP1, IL-1β, and IL-6 mRNA levels in lipid mixture (LM)-induced hepatocytes in a dose-dependent manner. These changes were reversed by 3-MA, an autophagy inhibitor. CONCLUSION Our findings suggest that canagliflozin ameliorates the pathogenesis of NAFLD by regulating lipid metabolism and inhibiting inflammation, which may be associated with its promotion of autophagy.
Collapse
Affiliation(s)
- Zhipeng Xu
- Department of Urology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Organ Transplantation and Nephrosis, Shandong Institute of Nephrology, Jinan, Shandong, China
| | - Wenxin Hu
- Department of Urology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Organ Transplantation and Nephrosis, Shandong Institute of Nephrology, Jinan, Shandong, China
| | - Bin Wang
- Department of Breast and Thyroid Surgery, Tengzhou Central People's Hospital, Zaozhuang, Shandong, China
| | - Ting Xu
- Department of Urology, Weifang Medical University, Weifang, Shandong, China
| | - Jianning Wang
- Department of Urology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Organ Transplantation and Nephrosis, Shandong Institute of Nephrology, Jinan, Shandong, China
| | - Dan Wei
- Department of Comprehensive Internal Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Institute of Nephrology, Jinan, Shandong, China.
| |
Collapse
|
13
|
Kang X, Amevor FK, Zhang L, Shah AM, Zhu Q, Tian Y, Shu G, Wang Y, Zhao X. Study on the Major Genes Related with Fat Deposition in Liver and Abdominal Fat of Different Breeds of Chicken. BRAZILIAN JOURNAL OF POULTRY SCIENCE 2022. [DOI: 10.1590/1806-9061-2020-1373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- X Kang
- Sichuan Agricultural University, China
| | - FK Amevor
- Sichuan Agricultural University, China
| | - L Zhang
- Sichuan Agricultural University, China
| | - AM Shah
- Sichuan Agricultural University, China
| | - Q Zhu
- Sichuan Agricultural University, China
| | - Y Tian
- Sichuan Agricultural University, China
| | - G Shu
- Sichuan Agricultural University, China
| | - Y Wang
- Sichuan Agricultural University, China
| | - X Zhao
- Sichuan Agricultural University, China
| |
Collapse
|
14
|
Chen S, Cai R, Liu Z, Cui H, She Z. Secondary metabolites from mangrove-associated fungi: source, chemistry and bioactivities. Nat Prod Rep 2021; 39:560-595. [PMID: 34623363 DOI: 10.1039/d1np00041a] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Covering 1989 to 2020The mangrove forests are a complex ecosystem occurring at tropical and subtropical intertidal estuarine zones and nourish a diverse group of microorganisms including fungi, actinomycetes, bacteria, cyanobacteria, algae, and protozoa. Among the mangrove microbial community, mangrove associated fungi, as the second-largest ecological group of the marine fungi, not only play an essential role in creating and maintaining this biosphere but also represent a rich source of structurally unique and diverse bioactive secondary metabolites, attracting significant attention of organic chemists and pharmacologists. This review summarizes the discovery relating to the source and characteristics of metabolic products isolated from mangrove-associated fungi over the past thirty years (1989-2020). Its emphasis included 1387 new metabolites from 451 papers, focusing on bioactivity and the unique chemical diversity of these natural products.
Collapse
Affiliation(s)
- Senhua Chen
- School of Chemistry, Sun Yat-sen University, Guangzhou 510006, China. .,School of Marine Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Runlin Cai
- School of Chemistry, Sun Yat-sen University, Guangzhou 510006, China. .,College of Science, Shantou University, Shantou 515063, China
| | - Zhaoming Liu
- School of Chemistry, Sun Yat-sen University, Guangzhou 510006, China. .,State Key Laboratory of Applied Microbiology Southern China, Guangdong Guangdong Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Hui Cui
- School of Chemistry, Sun Yat-sen University, Guangzhou 510006, China. .,School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Zhigang She
- School of Chemistry, Sun Yat-sen University, Guangzhou 510006, China.
| |
Collapse
|
15
|
Liang B, Cai XY, Gu N. Marine Natural Products and Coronary Artery Disease. Front Cardiovasc Med 2021; 8:739932. [PMID: 34621803 PMCID: PMC8490644 DOI: 10.3389/fcvm.2021.739932] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 08/24/2021] [Indexed: 12/18/2022] Open
Abstract
Coronary artery disease is the major cause of mortality worldwide, especially in low- and middle-income earners. To not only reduce angina symptoms and exercise-induced ischemia but also prevent cardiovascular events, pharmacological intervention strategies, including antiplatelet drugs, anticoagulant drugs, statins, and other lipid-lowering drugs, and renin-angiotensin-aldosterone system blockers, are conducted. However, the existing drugs for coronary artery disease are incomprehensive and have some adverse reactions. Thus, it is necessary to look for new drug research and development. Marine natural products have been considered a valuable source for drug discovery because of their chemical diversity and biological activities. The experiments and investigations indicated that several marine natural products, such as organic small molecules, polysaccharides, proteins, and bioactive peptides, and lipids were effective for treating coronary artery disease. Here, we particularly discussed the functions and mechanisms of active substances in coronary artery disease, including antiplatelet, anticoagulant, lipid-lowering, anti-inflammatory, and antioxidant activities.
Collapse
Affiliation(s)
- Bo Liang
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Xin-Yi Cai
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Ning Gu
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
16
|
Gong H, Bandura J, Wang GL, Feng ZP, Sun HS. Xyloketal B: A marine compound with medicinal potential. Pharmacol Ther 2021; 230:107963. [PMID: 34375691 DOI: 10.1016/j.pharmthera.2021.107963] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 06/01/2021] [Accepted: 07/13/2021] [Indexed: 12/14/2022]
Abstract
In recent decades, technological advantages have allowed scientists to isolate medicinal compounds from marine organisms that exhibit unique structure and bioactivity. The mangrove fungus Xylaria sp. from the South China Sea is rich in metabolites and produces a potent therapeutic compound, xyloketal B. Since its isolation in 2001, xyloketal B has been extensively studied in a wide variety of cell types and in vitro and in vivo disease models. Xyloketal B and its derivatives exhibit cytoprotective effects in cardiovascular and neurodegenerative diseases by reducing oxidative stress, regulating the apoptosis pathway, maintaining ionic balance, mitigating inflammatory responses, and preventing protein aggregation. Xyloketal B has also shown to alleviate lipid accumulation in a non-alcoholic fatty liver disease model. Moreover, xyloketal B treatment induces glioblastoma cell death. This review summarizes our current understanding of xyloketal B in various disease models.
Collapse
Affiliation(s)
- Haifan Gong
- Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada; Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Julia Bandura
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Guan-Lei Wang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China; Key Laboratory of Functional Molecules from Oceanic Microorganisms (Sun Yat-Sen University), Department of Education of Guangdong Province, 510080, China.
| | - Zhong-Ping Feng
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada.
| | - Hong-Shuo Sun
- Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada; Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada; Department of Pharmacology and Toxicology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada; Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada.
| |
Collapse
|
17
|
Wei D, Wu S, Liu J, Zhang X, Guan X, Gao L, Xu Z. Theobromine ameliorates nonalcoholic fatty liver disease by regulating hepatic lipid metabolism via mTOR signaling pathway in vivo and in vitro. Can J Physiol Pharmacol 2021; 99:775-785. [PMID: 33290156 DOI: 10.1139/cjpp-2020-0259] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Theobromine, a methylxanthine present in cocoa, has been shown to possess many beneficial pharmacological properties such as anti-oxidative stress, anti-inflammatory property, and anti-microbial activity. In this study, we investigated the effects of theobromine on nonalcoholic fatty liver disease (NAFLD) and the possible underlying mechanisms in vivo and in vitro. The results showed that theobromine reduced body weight and fat mass and improved dyslipidemia. Theobromine mitigated liver injury and significantly reduced hepatic triglyceride level in mice with obesity. Histological examinations also showed hepatic steatosis was alleviated after theobromine treatment. Furthermore, theobromine reversed the elevated mRNA and protein expression of SREBP-1c, FASN, CD36, FABP4, and the suppressed expression of PPARα and CPT1a in the liver of mice with obesity, which were responsible for lipogenesis, fatty acid uptake, and fatty acid oxidation respectively. In vitro, theobromine also downregulated SREBP-1c, FASN, CD36, FABP4 and upregulated PPARα and CPT1a mRNA and protein levels in hepatocytes in a dose-dependent manner, while these changes were reversed by L-leucine, a mammalian target of rapamycin (mTOR) agonist. The present study demonstrated that theobromine improved NAFLD by inhibiting lipogenesis and fatty acid uptake and promoting fatty acid oxidation in the liver and hepatocytes, which might be associated with its suppression of mTOR signaling pathway. Novelty: Theobromine protects against high-fat diet - induced NAFLD. Theobromine inhibits lipogenesis and fatty acid uptake and promotes fatty acid oxidation in the liver and hepatocytes via inhibiting mTOR signaling pathway.
Collapse
Affiliation(s)
- Dan Wei
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Shaofei Wu
- Department of Hepatology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Jie Liu
- Department of Public Health, Tengzhou Central People's Hospital, Zaozhuang, Shandong, China
| | - Xiaoqian Zhang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Xiaoling Guan
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Li Gao
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Zhipeng Xu
- Department of Urology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
18
|
Mayer AMS, Guerrero AJ, Rodríguez AD, Taglialatela-Scafati O, Nakamura F, Fusetani N. Marine Pharmacology in 2016-2017: Marine Compounds with Antibacterial, Antidiabetic, Antifungal, Anti-Inflammatory, Antiprotozoal, Antituberculosis and Antiviral Activities; Affecting the Immune and Nervous Systems, and Other Miscellaneous Mechanisms of Action. Mar Drugs 2021; 19:49. [PMID: 33494402 PMCID: PMC7910995 DOI: 10.3390/md19020049] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 12/12/2022] Open
Abstract
The review of the 2016-2017 marine pharmacology literature was prepared in a manner similar as the 10 prior reviews of this series. Preclinical marine pharmacology research during 2016-2017 assessed 313 marine compounds with novel pharmacology reported by a growing number of investigators from 54 countries. The peer-reviewed literature reported antibacterial, antifungal, antiprotozoal, antituberculosis, and antiviral activities for 123 marine natural products, 111 marine compounds with antidiabetic and anti-inflammatory activities as well as affecting the immune and nervous system, while in contrast 79 marine compounds displayed miscellaneous mechanisms of action which upon further investigation may contribute to several pharmacological classes. Therefore, in 2016-2017, the preclinical marine natural product pharmacology pipeline generated both novel pharmacology as well as potentially new lead compounds for the growing clinical marine pharmaceutical pipeline, and thus sustained with its contributions the global research for novel and effective therapeutic strategies for multiple disease categories.
Collapse
Affiliation(s)
- Alejandro M. S. Mayer
- Department of Pharmacology, College of Graduate Studies, Midwestern University, 555 31st Street, Downers Grove, IL 60515, USA;
| | - Aimee J. Guerrero
- Department of Pharmacology, College of Graduate Studies, Midwestern University, 555 31st Street, Downers Grove, IL 60515, USA;
| | - Abimael D. Rodríguez
- Molecular Sciences Research Center, University of Puerto Rico, 1390 Ponce de León Avenue, San Juan, PR 00926, USA;
| | | | - Fumiaki Nakamura
- Department of Chemistry and Biochemistry, Graduate School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan;
| | | |
Collapse
|
19
|
Zhai M, Yan X, Liu J, Long Z, Zhao S, Li W, Liu Y, Hai C. Electromagnetic Fields Ameliorate Insulin Resistance and Hepatic Steatosis by Modulating Redox Homeostasis and SREBP-1c Expression in db/db Mice. Diabetes Metab Syndr Obes 2021; 14:1035-1042. [PMID: 33727836 PMCID: PMC7954280 DOI: 10.2147/dmso.s294020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 02/24/2021] [Indexed: 12/11/2022] Open
Abstract
PURPOSE The prevalence of nonalcoholic fatty liver disease (NAFLD), which has recently become known as metabolic-associated fatty liver disease (MAFLD), has risen. However, pharmacotherapies for this disease have not been approved. Electromagnetic fields (EMFs) have excellent bioeffects on multiple diseases. However, the effects of EMFs on NAFLD are unknown. This study investigated the bioeffects of EMF exposure on insulin resistance, liver redox homeostasis and hepatic steatosis in db/db mice. METHODS Animals were sacrificed after EMF exposure for 8 weeks. The fasting blood glucose and insulin levels in the serum were tested. The homeostatic model assessment of insulin resistance (HOMA-IR) was calculated by a formula. The levels of MDA, GSSG and GSH, biomarkers of redox, were assessed. The activities of CAT, SOD and GSH-Px were assessed. The body and liver weights were measured. Hepatic lipid accumulation was observed by Oil Red O staining. Hepatic CAT, GR, GSH-Px, SOD1, SOD2 and SREBP-1 expression was determined by Western blotting. RESULTS EMF exposure ameliorated insulin resistance and oxidative stress in the liver by downregulating the MDA and GSSG levels, increasing the reduced GSH levels, and promoting the GSH-Px levels in db/db mice. In addition, liver weight and triglyceride (TG) levels were reduced by EMF exposure. Simultaneously, EMF exposure improved hepatic steatosis by downregulating the protein expression of SREBP-1c. CONCLUSION The present findings suggest that EMF exposure has positive effects in the treatment of NAFLD.
Collapse
Affiliation(s)
- Mingming Zhai
- Department of Biomedical Engineering, Air Force Medical University, Xi’an, People’s Republic of China
| | - Xi Yan
- Department of Dermatology, The Second Affiliated Hospital, Air Force Medical University, Xi’an, People’s Republic of China
| | - Jiangzheng Liu
- Department of Toxicology, Shanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University, Xi’an, People’s Republic of China
| | - Zi Long
- Department of Toxicology, Shanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University, Xi’an, People’s Republic of China
| | - Siyan Zhao
- Institute of Nuclear Biological and Chemical Defence, Beijing, People’s Republic of China
| | - Wendan Li
- Institute of Nuclear Biological and Chemical Defence, Beijing, People’s Republic of China
| | - Ying Liu
- Institute of Nuclear Biological and Chemical Defence, Beijing, People’s Republic of China
- Ying Liu Institute of Nuclear Biological and Chemical Defence, No. 1, Yangfang Zhongxin North Street, Beijing, 102205, People’s Republic of China Email
| | - Chunxu Hai
- Department of Toxicology, Shanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University, Xi’an, People’s Republic of China
- Correspondence: Chunxu Hai Air Force Medical University (AFMU), No. 169 Changle West Road, Xi’an, Shaanxi, 710032, People’s Republic of ChinaTel +86-29-84774879 Email
| |
Collapse
|
20
|
Cheng J, Liu Y, Liu Y, Liu D, Liu Y, Guo Y, Wu Z, Li H, Wang H. Ursolic acid alleviates lipid accumulation by activating the AMPK signaling pathway in vivo and in vitro. J Food Sci 2020; 85:3998-4008. [PMID: 33001454 DOI: 10.1111/1750-3841.15475] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 08/10/2020] [Accepted: 09/08/2020] [Indexed: 02/06/2023]
Abstract
The mechanism underlying the effect of ursolic acid (UA) on lipid metabolism remains unclear. This study aimed to explore the mechanisms of UA in reducing lipid accumulation in free fatty acids-cultured HepG2 cells and in high-fat-diet-fed C57BL/6J mice. In vivo, UA effectively alleviated liver steatosis and decreased the size of adipocytes in the epididymis. It also significantly decreased the total cholesterol (TC) and triglyceride (TG) contents in the liver and plasma in C57BL/6 mice. In vitro, UA (20 µM) significantly reduced lipid accumulation; the intracellular TC contents decreased from 0.078 ± 0.0047 to 0.049 ± 0.0064 µmol/mg protein, and TG contents from 0.133 ± 0.005 to 0.066 ± 0.0047 µmol/mg protein, in HepG2 cells. Furthermore, UA reduced the mRNA expression related to fat synthesis, enhanced the mRNA expression related to adipose decomposition, and dramatically upregulated the protein expression of P-AMPK in vivo and in vitro. Of note, these protective effects of UA on a high-fat environment were blocked by the AMPK inhibitor (compound C) in vitro. In addition, the molecular docking results suggested that UA could be docked to the AMPK protein as an AMPK activator. These results indicated that UA lowered the lipid content probably via activating the AMPK signaling pathway, thereby inhibiting lipid synthesis and promoting fat decomposition. PRACTICAL APPLICATION: Ursolic acid (UA) widely exists in vegetables and fruits. This study highlighted a lipid-lowing mechanism of UA in HepG2 cells and C57BL/6J mice. The data indicated that UA might be used in lipid-lowering functional foods.
Collapse
Affiliation(s)
- Jing Cheng
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology (TUST), Tianjin, 300457, China
| | - Ying Liu
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology (TUST), Tianjin, 300457, China
| | - Yaojie Liu
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology (TUST), Tianjin, 300457, China
| | - Dong Liu
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology (TUST), Tianjin, 300457, China
| | - Yang Liu
- Animal and Plant and Food Inspection Center of Tianjin Customs (Former Tianjin Inspection and Quarantine Bureau), Tianjin, 300461, China
| | - Yatu Guo
- Tianjin Eye Hospital, Tianjin Eye Institute, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin, 300384, China
| | - Zijian Wu
- College of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin, 300143, China
| | - Heyu Li
- Tianjin Ubasio Biotechnology Group Co. Ltd., Tianjin, 300457, China
| | - Hao Wang
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology (TUST), Tianjin, 300457, China
| |
Collapse
|
21
|
Balkrishna A, Gohel V, Singh R, Joshi M, Varshney Y, Srivastava J, Bhattacharya K, Varshney A. Tri-Herbal Medicine Divya Sarva-Kalp-Kwath (Livogrit) Regulates Fatty Acid-Induced Steatosis in Human HepG2 Cells through Inhibition of Intracellular Triglycerides and Extracellular Glycerol Levels. Molecules 2020; 25:molecules25204849. [PMID: 33096687 PMCID: PMC7587968 DOI: 10.3390/molecules25204849] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/09/2020] [Accepted: 10/13/2020] [Indexed: 12/20/2022] Open
Abstract
Steatosis is characterized by excessive triglycerides accumulation in liver cells. Recently, application of herbal formulations has gained importance in treating complex diseases. Therefore, this study explores the efficacy of tri-herbal medicine Divya Sarva-Kalp-Kwath (SKK; brand name, Livogrit) in treating free fatty acid (FFA)-induced steatosis in human liver (HepG2) cells and rat primary hepatocytes. Previously, we demonstrated that cytosafe SKK ameliorated CCl4-induced hepatotoxicity. In this study, we evaluated the role of SKK in reducing FFA-induced cell-death, and steatosis in HepG2 through analysis of cell viability, intracellular lipid and triglyceride accumulation, extracellular free glycerol levels, and mRNA expression changes. Plant metabolic components fingerprinting in SKK was performed via High Performance Thin Layer Chromatography (HPTLC). Treatment with SKK significantly reduced the loss of cell viability induced by 2 mM-FFA in a dose-dependent manner. SKK also reduced intracellular lipid, triglyceride accumulation, secreted AST levels, and increased extracellular free glycerol presence in the FFA-exposed cells. SKK normalized the FFA-stimulated overexpression of SREBP1c, FAS, C/EBPα, and CPT1A genes associated with the induction of steatosis. In addition, treatment of rat primary hepatocytes with FFA and SKK concurrently, reduced intracellular lipid accumulation. Thus, SKK showed efficacy in reducing intracellular triglyceride accumulation and increasing extracellular glycerol release, along with downregulation of related key genetic factors for FFA-associated steatosis.
Collapse
Affiliation(s)
- Acharya Balkrishna
- Drug Discovery and Development Division, Patanjali Research Institute, Governed by Patanjali Research Foundation Trust, NH-58, Haridwar 249 405, Uttarakhand, India; (A.B.); (V.G.); (R.S.); (M.J.); (Y.V.); (J.S.); (K.B.)
- Department of Allied and Applied Sciences, University of Patanjali, Patanjali Yog Peeth, Roorkee-Haridwar Road, Haridwar 249 405, Uttarakhand, India
- Patanjali Yog Peeth (UK) Trust, 40 Lambhill Street, Kinning Park, Glasgow G41 1AU, UK
| | - Vivek Gohel
- Drug Discovery and Development Division, Patanjali Research Institute, Governed by Patanjali Research Foundation Trust, NH-58, Haridwar 249 405, Uttarakhand, India; (A.B.); (V.G.); (R.S.); (M.J.); (Y.V.); (J.S.); (K.B.)
| | - Rani Singh
- Drug Discovery and Development Division, Patanjali Research Institute, Governed by Patanjali Research Foundation Trust, NH-58, Haridwar 249 405, Uttarakhand, India; (A.B.); (V.G.); (R.S.); (M.J.); (Y.V.); (J.S.); (K.B.)
| | - Monali Joshi
- Drug Discovery and Development Division, Patanjali Research Institute, Governed by Patanjali Research Foundation Trust, NH-58, Haridwar 249 405, Uttarakhand, India; (A.B.); (V.G.); (R.S.); (M.J.); (Y.V.); (J.S.); (K.B.)
| | - Yash Varshney
- Drug Discovery and Development Division, Patanjali Research Institute, Governed by Patanjali Research Foundation Trust, NH-58, Haridwar 249 405, Uttarakhand, India; (A.B.); (V.G.); (R.S.); (M.J.); (Y.V.); (J.S.); (K.B.)
| | - Jyotish Srivastava
- Drug Discovery and Development Division, Patanjali Research Institute, Governed by Patanjali Research Foundation Trust, NH-58, Haridwar 249 405, Uttarakhand, India; (A.B.); (V.G.); (R.S.); (M.J.); (Y.V.); (J.S.); (K.B.)
| | - Kunal Bhattacharya
- Drug Discovery and Development Division, Patanjali Research Institute, Governed by Patanjali Research Foundation Trust, NH-58, Haridwar 249 405, Uttarakhand, India; (A.B.); (V.G.); (R.S.); (M.J.); (Y.V.); (J.S.); (K.B.)
| | - Anurag Varshney
- Drug Discovery and Development Division, Patanjali Research Institute, Governed by Patanjali Research Foundation Trust, NH-58, Haridwar 249 405, Uttarakhand, India; (A.B.); (V.G.); (R.S.); (M.J.); (Y.V.); (J.S.); (K.B.)
- Department of Allied and Applied Sciences, University of Patanjali, Patanjali Yog Peeth, Roorkee-Haridwar Road, Haridwar 249 405, Uttarakhand, India
- Correspondence: ; Tel.: +91-1334-244-107 (ext. x7458); Fax: +91-1334-244-805
| |
Collapse
|
22
|
Roy B, Rout N, Kuila P, Sarkar D. Synthesis and structural anomaly of
xyloketals‐unique
benzoxacycles: A review. J Heterocycl Chem 2020. [DOI: 10.1002/jhet.4152] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Barnali Roy
- Department of Chemistry NIT Rourkela Odisha India
| | | | | | | |
Collapse
|
23
|
Zhang AR, Sun J, He Y, Wang N, Tian L. Attenuation of lipid accumulation in Bel-7402 cells through ADPN/AMPKα signaling stimulated by Fructus rosae laxae extract. J Food Biochem 2020; 44:e13497. [PMID: 33029846 DOI: 10.1111/jfbc.13497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 09/05/2020] [Accepted: 09/09/2020] [Indexed: 12/29/2022]
Abstract
In this work, a comparison study was conducted on the contents of total flavonoids and hyperoside in different polarity extracts of Fructus rosae laxae (FRL). The lipid-lowering effect and mechanism of FRL ethyl acetate extract (FRLE) on the lipid accumulation model of Bel-7402 cells in vitro were studied. The results showed that the contents of total flavonoids and hyperoside in FRLE were significantly higher than those in the other polarity extracts. Compared with those in the model group, the levels of triglyceride and total cholesterol decreased, the activities of superoxide dismutase and lactate dehydrogenase increased, and the levels of inflammatory factors interleukin-6 and tumor necrosis factor-α decreased significantly in the cells intervened with FRLE. Moreover, FRLE can regulate lipid metabolism by activating the AMP-activated protein kinase α phosphorylation pathway and increasing the expression of adiponectin. PRACTICAL APPLICATIONS: Fructus rosae laxae (FRL) is an edible medicinal fruit with multiple biological activities, such as antioxidation, anti-inflammatory, and hepatoprotective properties. However, the lipid-lowering activity of FRL and its mechanism of action have not yet been investigated. Our data indicate that the FRL extract, which contains high levels of antioxidant and anti-inflammatory components, plays a beneficial role in regulating lipid metabolism disorders, mainly by regulating the expression of proteins involved in the ADPN/AMPK signaling pathway, and reduces the release of inflammatory factors. Thus, the FRL extract effectively reduces the accumulation of free fatty acids (FFA) in vitro and exhibits considerable potential for the prevention and treatment lipid metabolism disorders.
Collapse
Affiliation(s)
- Ai-Rong Zhang
- College of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, P.R.China
| | - Jing Sun
- College of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, P.R.China
| | - Yuan He
- College of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, P.R.China
| | - Ning Wang
- College of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, P.R.China
| | - Li Tian
- College of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, P.R.China
| |
Collapse
|
24
|
Zhao J, Cao Q, Xing M, Xiao H, Cheng Z, Song S, Ji A. Advances in the Study of Marine Products with Lipid-Lowering Properties. Mar Drugs 2020; 18:E390. [PMID: 32726987 PMCID: PMC7459887 DOI: 10.3390/md18080390] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 12/18/2022] Open
Abstract
With twice the number of cancer's deaths, cardiovascular diseases have become the leading cause of death worldwide. Atherosclerosis, in particular, is a progressive, chronic inflammatory cardiovascular disease caused by persistent damage to blood vessels due to elevated cholesterol levels and hyperlipidemia. This condition is characterized by an increase in serum cholesterol, triglycerides, and low-density lipoprotein, and a decrease in high-density lipoprotein. Although existing therapies with hypolipidemic effects can improve the living standards of patients with cardiovascular diseases, the drugs currently used in clinical practice have certain side effects, which insists on the need for the development of new types of drugs with lipid-lowering effects. Some marine-derived substances have proven hypolipidemic activities with fewer side effects and stand as a good alternative for drug development. Recently, there have been thousands of studies on substances with lipid-lowering properties of marine origin, and some are already implemented in clinical practice. Here, we summarize the active components of marine-derived products having a hypolipidemic effect. These active constituents according to their source are divided into algal, animal, plant and microbial and contribute to the development and utilization of marine medicinal products with hypolipidemic effects.
Collapse
Affiliation(s)
- Jiarui Zhao
- Marine College, Shandong University, Weihai 264209, China; (J.Z.); (Q.C.); (M.X.); (H.X.); (Z.C.)
| | - Qi Cao
- Marine College, Shandong University, Weihai 264209, China; (J.Z.); (Q.C.); (M.X.); (H.X.); (Z.C.)
| | - Maochen Xing
- Marine College, Shandong University, Weihai 264209, China; (J.Z.); (Q.C.); (M.X.); (H.X.); (Z.C.)
| | - Han Xiao
- Marine College, Shandong University, Weihai 264209, China; (J.Z.); (Q.C.); (M.X.); (H.X.); (Z.C.)
| | - Zeyu Cheng
- Marine College, Shandong University, Weihai 264209, China; (J.Z.); (Q.C.); (M.X.); (H.X.); (Z.C.)
| | - Shuliang Song
- Marine College, Shandong University, Weihai 264209, China; (J.Z.); (Q.C.); (M.X.); (H.X.); (Z.C.)
| | - Aiguo Ji
- Marine College, Shandong University, Weihai 264209, China; (J.Z.); (Q.C.); (M.X.); (H.X.); (Z.C.)
- School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| |
Collapse
|
25
|
Li H, Xi Y, Xin X, Tian H, Hu Y. Gypenosides regulate farnesoid X receptor-mediated bile acid and lipid metabolism in a mouse model of non-alcoholic steatohepatitis. Nutr Metab (Lond) 2020; 17:34. [PMID: 32377219 PMCID: PMC7195801 DOI: 10.1186/s12986-020-00454-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 04/22/2020] [Indexed: 02/07/2023] Open
Abstract
Background Gypenosides (Gyp) are the main ingredient of the Chinese medicine, Gynostemma pentaphyllum. They are widely used in Asia as a hepatoprotective agent. Here, we elucidated the mechanism of Gyp in non-alcoholic steatohepatitis (NASH) with a focus on farnesoid X receptor (FXR)-mediated bile acid and lipid metabolic pathways. Methods NASH was induced in mice by high-fat diet (HFD) feeding, while mice in the control group were given a normal diet. At the end of week 10, HFD-fed mice were randomly divided into HFD, HFD plus Gyp, and HFD plus obeticholic acid (OCA, FXR agonist) groups and were given the corresponding treatments for 4 weeks. Next, we analyzed the histopathological changes as well as the liver triglyceride (TG) level and serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), fasting blood glucose (FBG), fasting insulin (FINS), TG, total cholesterol (TC), and low-density lipoprotein cholesterol (LDL-C) levels as well as the bile acid profile. We carried out RT-PCR and western blotting to detect HFD-induced alterations in gene/protein expression related to bile acid and lipid metabolism. Results The HFD group had histopathological signs of hepatic steatosis and vacuolar degeneration. The liver TG and serum ALT, AST, FBG, FINS, TC, and LDL-C levels as well as the total bile acid level were significantly higher in the HFD group than in the control group (P < 0.01). In addition, we observed significant changes in the expression of proteins involved in bile acid or lipid metabolism (P < 0.05). Upon treatment with Gyp or OCA, signs of hepatic steatosis and alterations in different biochemical parameters were significantly improved (P < 0.05). Further, HFD-induced alterations in the expression genes involved in bile acid and lipid metabolism, such as CYP7A1, BSEP, SREBP1, and FASN, were significantly alleviated. Conclusions Gyp can improve liver lipid and bile acid metabolism in a mouse model of NASH, and these effects may be related to activation of the FXR signaling pathway.
Collapse
Affiliation(s)
- Hongshan Li
- 1Institute of Liver Disease, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Liver Disease Department, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang China.,Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo, 315010 Zhejiang China
| | - Yingfei Xi
- 4Medical School of Ningbo University, Ningbo, Zhejiang China
| | - Xin Xin
- 1Institute of Liver Disease, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huajie Tian
- 1Institute of Liver Disease, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiyang Hu
- 1Institute of Liver Disease, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
26
|
Wang J, He W, Tsai PJ, Chen PH, Ye M, Guo J, Su Z. Mutual interaction between endoplasmic reticulum and mitochondria in nonalcoholic fatty liver disease. Lipids Health Dis 2020; 19:72. [PMID: 32284046 PMCID: PMC7155254 DOI: 10.1186/s12944-020-01210-0] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 02/24/2020] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a common metabolic syndrome. Imbalances between liver lipid output and input are the direct causes of NAFLD, and hepatic steatosis is the pathological premise and basis for NAFLD progression. Mutual interaction between endoplasmic reticulum stress (ERS) and oxidative stress play important roles in NAFLD pathogenesis. Notably, mitochondria-associated membranes (MAMs) act as a structural bridges for functional clustering of molecules, particularly for Ca2+, lipids, and reactive oxygen species (ROS) exchange. Previous studies have examined the crucial roles of ERS and ROS in NAFLD and have shown that MAM structural and functional integrity determines normal ER- mitochondria communication. Upon disruption of MAM integrity, miscommunication directly or indirectly causes imbalances in Ca2+ homeostasis and increases ERS and oxidative stress. Here, we emphasize the involvement of MAMs in glucose and lipid metabolism, chronic inflammation and insulin resistance in NAFLD and summarize MAM-targeting drugs and compounds, most of which achieve their therapeutic or ameliorative effects on NAFLD by improving MAM integrity. Therefore, targeting MAMs may be a viable strategy for NAFLD treatment. This review provides new ideas and key points for basic NAFLD research and drug development centred on mitochondria and the endoplasmic reticulum.
Collapse
Affiliation(s)
- Jin Wang
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, 510006, China.,Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong TCM Key Laboratory for Metabolic Diseases, Key Laboratory of Modulating Liver to Treat Hyperlipemia SATCM, Level 3 Laboratory of Lipid Metabolism SATCM, Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Wanping He
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, 510006, China.,Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong TCM Key Laboratory for Metabolic Diseases, Key Laboratory of Modulating Liver to Treat Hyperlipemia SATCM, Level 3 Laboratory of Lipid Metabolism SATCM, Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Ping-Ju Tsai
- King-Prebiotics Biotechnology (TW) Co., LTD, 2F.-1, No. 250, Zhongshan Rd., Linkou Dist, New Taipei City, 24446, Taiwan
| | - Pei-Hsuan Chen
- King-Prebiotics Biotechnology (TW) Co., LTD, 2F.-1, No. 250, Zhongshan Rd., Linkou Dist, New Taipei City, 24446, Taiwan
| | - Manxiang Ye
- New Francisco (Yunfu City) Biotechnology Co, Ltd Swan-kan-chiau Ind. Dist., Kaofong Village, Yunfu City, Guangdong, China
| | - Jiao Guo
- Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong TCM Key Laboratory for Metabolic Diseases, Key Laboratory of Modulating Liver to Treat Hyperlipemia SATCM, Level 3 Laboratory of Lipid Metabolism SATCM, Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Zhengquan Su
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| |
Collapse
|
27
|
Cheng J, Liu D, Zhao J, Li X, Yan Y, Wu Z, Wang H, Wang C. Lutein attenuates oxidative stress and inhibits lipid accumulation in free fatty acids-induced HepG2 cells by activating the AMPK pathway. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.103445] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
28
|
Liou CJ, Dai YW, Wang CL, Fang LW, Huang WC. Maslinic acid protects against obesity-induced nonalcoholic fatty liver disease in mice through regulation of the Sirt1/AMPK signaling pathway. FASEB J 2019; 33:11791-11803. [PMID: 31361524 DOI: 10.1096/fj.201900413rrr] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Maslinic acid is a pentacyclic triterpenoid that is distributed in the peel of olives. Previous studies found that maslinic acid inhibited inflammatory response and antioxidant effects. We investigated whether maslinic acid ameliorates nonalcoholic fatty liver disease in mice with high-fat-diet (HFD)-induced obesity and evaluated the regulation of lipogenesis in hepatocytes. Male C57BL/6 mice fed a normal diet or HFD (60% fat, w/w) were tested for 16 wk. After the fourth week, mice were injected intraperitoneally with maslinic acid for 12 wk. In another experiment, HepG2 cells were treated with oleic acid to induce lipid accumulation or maslinic acid to evaluate lipogenesis. Maslinic acid significantly reduced body weight compared with HFD-fed mice. Maslinic acid reduced liver weight and liver lipid accumulation and improved hepatocyte steatosis. Furthermore, serum glucose, leptin, and free fatty acid concentrations significantly reduced, but the serum adiponectin concentration was higher, in the maslinic acid group than in the HFD group. In liver tissue, maslinic acid suppressed transcription factors involved in lipogenesis and increased adipose triglyceride lipase. In vitro, maslinic acid decreased lipogenesis by activating AMPK. These findings suggest that maslinic acid acts against hepatic steatosis by regulating enzyme activity involved in lipogenesis, lipolysis, and fatty acid oxidation in the liver.-Liou, C.-J., Dai, Y.-W., Wang, C.-L., Fang, L.-W., Huang, W.-C. Maslinic acid protects against obesity-induced nonalcoholic fatty liver disease in mice through regulation of the Sirt1/AMPK signaling pathway.
Collapse
Affiliation(s)
- Chian-Jiun Liou
- Division of Basic Medical Sciences, Department of Nursing, Research Center for Chinese Herbal Medicine, Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, Taoyuan City, Taiwan.,Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Taoyuan City, Taiwan
| | - Yi-Wen Dai
- Research Center for Food and Cosmetic Safety, Research Center for Chinese Herbal Medicine, Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan City, Taiwan
| | - Chia-Ling Wang
- Research Center for Food and Cosmetic Safety, Research Center for Chinese Herbal Medicine, Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan City, Taiwan
| | - Li-Wen Fang
- Department of Nutrition, I-Shou University, Kaohsiung City, Taiwan
| | - Wen-Chung Huang
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Taoyuan City, Taiwan.,Research Center for Food and Cosmetic Safety, Research Center for Chinese Herbal Medicine, Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan City, Taiwan
| |
Collapse
|
29
|
Lei Y, Gong L, Tan F, Liu Y, Li S, Shen H, Zhu M, Cai W, Xu F, Hou B, Zhou Y, Han H, Qiu L, Sun H. Vaccarin ameliorates insulin resistance and steatosis by activating the AMPK signaling pathway. Eur J Pharmacol 2019; 851:13-24. [DOI: 10.1016/j.ejphar.2019.02.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 02/14/2019] [Accepted: 02/15/2019] [Indexed: 12/26/2022]
|
30
|
Yu Z, Mao C, Fu X, Ma M. High Density Lipoprotein from Egg Yolk (EYHDL) Improves Dyslipidemia by Mediating Fatty Acids Metabolism in High Fat Diet-induced Obese Mice. Food Sci Anim Resour 2019; 39:179-196. [PMID: 31149661 PMCID: PMC6533406 DOI: 10.5851/kosfa.2018.e38] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 09/14/2018] [Accepted: 09/17/2018] [Indexed: 12/30/2022] Open
Abstract
We investigated the effect of high density lipoprotein from egg yolk (EYHDL) on
serum, hepatic and fecal lipid and fatty acids (FAs) levels and on gene
expression involved in FAs metabolism. Male KM mice were fed either normal diet
(ND; n=20), high fat diet (HFD; n=20), or high fat diet containing
EYHDL (EYHDL; 0.6 mg/g, every day by oral gavage, n=20) for 100 days. At
the end of the experiment, the effects of treatments on biochemical parameters,
FAs profiles and involved gene expression were analyzed. Our results revealed
that EYHDL markedly suppressed the body weight gain, accumulation of abdominal
fat tissues, serum concentrations of LDL-cholesterol (LDL-C) and triglycerides,
hepatic triglycerides and cholesterol accumulation, while increased serum
concentration of HDL-cholesterol (HDL-C). EYHDL intake also increased total
cholesterol (TC) excretions compared with HFD group. Moreover, it alleviated the
severity of fatty liver and improved glucose and insulin tolerance compared with
HFD. More importantly, EYHDL partially normalized FAs profiles in serum, liver
and fecaces and neutralized the HFD-induced upregulation of SREBP-1c, Acaca,
Fasn, GPAT and Scd1. In conclusion, our findings indicate that EYHDL may have
the potential to improve metabolic disturbances that occur in HFD mice and can
be considered as an appropriate dietary recommendation for the treatment of
metabolic syndrome (MetS).
Collapse
Affiliation(s)
- Zhihui Yu
- National R&D Center for Egg Processing, College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Changyi Mao
- National R&D Center for Egg Processing, College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Xing Fu
- National R&D Center for Egg Processing, College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Meihu Ma
- National R&D Center for Egg Processing, College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| |
Collapse
|
31
|
El-Sherbiny M, Eldosoky M, El-Shafey M, Othman G, Elkattawy HA, Bedir T, Elsherbiny NM. Vitamin D nanoemulsion enhances hepatoprotective effect of conventional vitamin D in rats fed with a high-fat diet. Chem Biol Interact 2018; 288:65-75. [PMID: 29653100 DOI: 10.1016/j.cbi.2018.04.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 03/27/2018] [Accepted: 04/10/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is associated with hyperlipidemia, obesity and type II diabetes. Due to increasing prevalence of these diseases globally, NAFLD is considered as a common form of chronic liver diseases. Vitamin D is a fat soluble vitamin with reported anti-inflammatory, anti-oxidant and immune modulating activity. Hypovitaminosis D often coexists with NAFLD and various studies reported beneficial role of vitamin D in modulating NAFLD. However, variable oral bioavailability, poor water solubility, and chemical degradation hinder the clinical application of vitamin D. PURPOSE We evaluated the potential protective effect of Vitamin D nanoemulsion (developed by sonication and pH-Shifting of pea protein isolate and canola oil) compared to conventional vitamin D against liver injury in rats fed with high fat diet (HFD). METHODS We analyzed liver function enzymes, lipid profile, lipid metabolism, levels and histopathology of inflammation and fibrosis in rat liver tissues. RESULTS HFD fed rats exhibited deterioration of liver function, poor lipid profile, decreased fatty acid oxidation and up-regulation of inflammatory cytokines and extracellular matrix deposition. Vitamin D administration reduced elevated liver enzymes, improved lipid profile, enhanced fatty acid oxidation and attenuated liver inflammation and fibrosis. Interestingly, vitamin D nanoemulsion was superior to conventional vitamin D with remarkable hepatoprotective effect against HFD-induced liver injury. CONCLUSION This study demonstrated vitamin D nanoemulsion as a more efficient formulation with more prominent hepatoprotective effect against HFD-induced liver injury compared to conventional oral vitamin D.
Collapse
Affiliation(s)
- Mohamed El-Sherbiny
- Anatomy Department, Mansoura Faculty of Medicine, Egypt; Almaarefa College of Medicine, Riyadh, Saudi Arabia
| | - Mohamed Eldosoky
- Medical Physiology Department, Mansoura Faculty of Medicine, Egypt
| | - Mohamed El-Shafey
- Anatomy Department, Mansoura Faculty of Medicine, Egypt; Fakeeh College for Medical Sciences, Jeddah, Saudi Arabia
| | - Gamal Othman
- Medical Biochemistry Department, Mansoura Faculty of Medicine, Egypt; Almaarefa College of Medicine, Riyadh, Saudi Arabia
| | - Hany A Elkattawy
- Medical Physiology Department, Zagazig Obesity Management and Research Unit, Zagazig Faculty of Medicine, Egypt; Almaarefa College of Medicine, Riyadh, Saudi Arabia
| | - Tamer Bedir
- Medical Microbiology and Immunology Department, Mansoura Faculty of Medicine, Egypt
| | - Nehal Mohsen Elsherbiny
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia; Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Egypt.
| |
Collapse
|