1
|
Ali MA, Michel HE, Menze ET, Tadros MG, Wahdan SA. The potential neuroprotective effect of empagliflozin against depressive-like behavior induced by chronic unpredictable mild stress in rats: Involvement of NLRP3 inflammasome. Eur J Pharmacol 2025; 998:177525. [PMID: 40107336 DOI: 10.1016/j.ejphar.2025.177525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 03/15/2025] [Accepted: 03/17/2025] [Indexed: 03/22/2025]
Abstract
Depression is a prevalent and debilitating condition that has a severe negative impact on a person's life. Chronic stress exposure plays a substantial role in the development of depression. In the present study, rats were exposed to chronic unpredictable mild stress (CUMS) for four weeks. Empagliflozin (EMPA), a Sodium-Glucose Cotransporter-2 (SGLT-2) inhibitor, is an oral antidiabetic agent exhibiting antioxidant, anti-inflammatory, and antiapoptotic effects. This study aimed to examine the antidepressant effect of EMPA in an experimental animal model of depression induced by CUMS in rats and explore the probable underlying mechanisms. Rats were treated with EMPA, per-orally, at a dose of 10 mg/kg/day for four weeks. EMPA treatment counteracted CUMS-induced histopathological, biochemical and behavioral alterations. EMPA suppressed the CUMS-induced increase in the oxidative stress, inflammatory, and apoptotic markers, where levels of MDA, IL-1β, TNF-α, NF-κB, NLRP3 and active caspase 3 were reduced by 29.6 %, 24.8 %, 17.9 %, 36.6 %, 24.5 % and 41.5 %, respectively, compared to the disease group. Furthermore, EMPA decreased the level of the microglial activation marker, iba-1 by 24 % in comparison to the disease group. In addition, EMPA treatment decreased blood glucose levels by 39 %, decreased serum insulin levels by 60.6 %, decreased HOMA-IR by 76.5 % and increased GLUT 4 expression, compared to the CUMS group, all which proves that EMPA has an effect insulin signaling and alleviates insulin resistance. Our results conclude that modulating key factors involved in depression, such as inflammation, oxidative stress, and NLRP3 inflammasome pathway, accounts for the anti-depressant effect of EMPA.
Collapse
Affiliation(s)
- Marwa A Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
| | - Haidy E Michel
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
| | - Esther T Menze
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
| | - Marianne G Tadros
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
| | - Sara A Wahdan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt.
| |
Collapse
|
2
|
Isah MB, Tajuddeen N, Yusuf A, Mohammed A, Ibrahim MA, Melzig M, Zhang X. The antidiabetic properties of lignans: a comprehensive review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156717. [PMID: 40220408 DOI: 10.1016/j.phymed.2025.156717] [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: 01/27/2025] [Revised: 03/23/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Diabetes mellitus (DM) is a chronic metabolic disease with a high global prevalence. Lignans, a class of plant natural compounds found in commonly consumed foods, are well-tolerated by humans and have demonstrated promising potential in the management of DM. Consumption of lignan-rich foods has been associated with improved overall health and quality of life. PURPOSE The clinical and preclinical evidence on the role of lignans in managing DM are critically examined. METHODS A thorough literature search was conducted across major scientific databases, focusing on studies that reported the effects of individual lignans on key diabetes indicators, such as glucose utilisation and insulin sensitivity, in both human and animal models, as well as in cell-based studies. RESULTS A total of 180 lignans were included in the review. Out of these, only three were investigated in randomised clinical trials in humans and 31 in animal models. The reviewed evidence suggests some beneficial effects of lignans in preventing the development of obesity-related diabetes. Their therapeutic benefits in preventing diabetes-related complications, particularly diabetic nephropathy, in both type 1 and type 2 diabetes, are also supported. Metabolites of various lignans, produced by microbial metabolism in the gut and serum enzymes, appear to be key bioactive forms, highlighting the need for detailed pharmacodynamic studies, optimised dosage designs, and the use of the appropriate lignan molecules for cell-based screening. CONCLUSION Lignans and their microbial metabolites show promise in preventing obesity-related diabetes and mitigating diabetes-related complications such as diabetic nephropathy, though further clinical studies are needed to optimize their therapeutic potential.
Collapse
Affiliation(s)
- Murtala Bindawa Isah
- Chinese-German Joint Laboratory for Natural Product Research, Shaanxi International Cooperation Demonstration Base, Shaanxi University of Technology, Hanzhong, 723000, Shaanxi, China; Department of Biochemistry, Umaru Musa Yar'adua University Katsina, Nigeria.
| | - Nasir Tajuddeen
- Department of Chemistry, Ahmadu Bello University Zaria, Nigeria
| | - Anas Yusuf
- Chinese-German Joint Laboratory for Natural Product Research, Shaanxi International Cooperation Demonstration Base, Shaanxi University of Technology, Hanzhong, 723000, Shaanxi, China
| | - Aminu Mohammed
- Department of Biochemistry, Ahmadu Bello University Zaria, Nigeria
| | | | - Matthias Melzig
- Chinese-German Joint Laboratory for Natural Product Research, Shaanxi International Cooperation Demonstration Base, Shaanxi University of Technology, Hanzhong, 723000, Shaanxi, China; Freie Universitaet Berlin, Institute of Pharmacy, Berlin, Germany.
| | - Xiaoying Zhang
- Chinese-German Joint Laboratory for Natural Product Research, Shaanxi International Cooperation Demonstration Base, Shaanxi University of Technology, Hanzhong, 723000, Shaanxi, China; Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, Campus de Gualtar, Braga, 4710-057, Portugal; Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada.
| |
Collapse
|
3
|
Chen TT, Yang JC, Chen GY, Dai YH, Zhang X, Chan HL, Lin TC, Wu YC. Silibinin, a PLC-β3 inhibitor, inhibits mast cell activation and alleviates OVA-induced asthma. Mol Immunol 2025; 178:76-86. [PMID: 39870013 DOI: 10.1016/j.molimm.2025.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/07/2025] [Accepted: 01/19/2025] [Indexed: 01/29/2025]
Abstract
The immunoglobulin E (IgE) receptor FcεRI (Fc epsilon RI) plays a crucial role in allergic reactions. Recent studies have indicated that the interaction between FcεRIβ and the downstream protein phospholipase C beta 3 (PLCβ3) leads to the production of inflammatory cytokines. The aim of this study was to develop small molecules that inhibit the protein-protein interactions between FcεRIβ and PLCβ3 to treat allergic inflammation. Additionally, PLCβ3 has emerged as a potential target protein for treating allergic inflammation. In this study, we employed a virtual screening technique to search the Taiwan Traditional Chinese Medicine Database, followed by a second screening using absorption, distribution, metabolism, excretion, and toxicity (ADMET). Among the compounds screened, silibinin exhibited the best performance, forming strong hydrogen bond interactions with residues of PLCβ3, with a binding free energy of -119.277 kcal/mol. Therefore, silibinin effectively blocked the interaction between FcεRIβ and PLCβ3. Silibinin reduced the production of allergic inflammatory cytokines, including cytokine-induced neutrophil chemoattractant 2a (CINC-2a), interleukin-2 (IL-2), cytokine-induced neutrophil chemoattractant 1 (CINC-1), interleukin 1α (IL-1α), macrophage inflammatory protein 3 alpha (MIP3α), interferon γ (IFN-γ), activin A, granulocyte macrophage colony stimulating factor (GM-CSF), intercellular adhesion molecule-1 (ICAM-1), interleukin 4 (IL-4), interleukin 13 (IL-13), Fas ligand (FasL) and tumor necrosis factor alpha (TNF-α), without inducing cytotoxicity. Furthermore, in studies of IgE-mediated allergic responses, silibinin also decreased the expression of surface IgE receptors (FcεRIs). Moreover, silibinin effectively alleviated allergen-induced asthma responses and reduced the infiltration of inflammatory immune cells into the lungs of an OVA-induced allergic airway inflammation mouse model. Taken together, these results demonstrate the potential antiallergic mechanism of silibinin both in vitro and in vivo, making it a promising candidate for the development of asthma therapeutics.
Collapse
Affiliation(s)
- Tzu-Ting Chen
- Institute of Bioinformatics and Structural Biology & Department of Medical Sciences, National Tsing Hua University, Hsinchu 300, Taiwan.
| | - Juan-Cheng Yang
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, Taiwan.
| | - Guan-Yu Chen
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, Taiwan.
| | - Yun-Hao Dai
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, Taiwan.
| | - Xiang Zhang
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm SE-17177, Sweden.
| | - Hong-Lin Chan
- Institute of Bioinformatics and Structural Biology & Department of Medical Sciences, National Tsing Hua University, Hsinchu 300, Taiwan.
| | - Tim Cc Lin
- Institute of Biomedical Science, National Chung-Hsing University, Taichung 402, Taiwan; Department of Medical Research, China Medical University Hospital, Taichung 404, Taiwan; Department of Medical Research, Taichung Veterans General Hospital, Taichung 407, Taiwan; Department of Pharmacology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Yang-Chang Wu
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, Taiwan; Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan; Master Program of Pharmaceutical Manufacture, College of Pharmacy, China Medical University, Taichung, Taiwan; Department of Medical Laboratory Science and Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan.
| |
Collapse
|
4
|
Huang Q, An Z, Xin X, Gou X, Tian X, Hu Y, Mei Z, Feng Q. The Effectiveness of Curcumin, Resveratrol, and Silymarin on MASLD: A Systematic Review and Meta-Analysis. Food Sci Nutr 2024; 12:10010-10029. [PMID: 39723101 PMCID: PMC11666838 DOI: 10.1002/fsn3.4595] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 09/25/2024] [Accepted: 10/25/2024] [Indexed: 12/28/2024] Open
Abstract
Polyphenols, known for their potent antioxidant and anti-inflammatory properties, have emerged as promising, natural, and safe complementary treatment options for metabolic-associated steatotic liver disease (MASLD). Among these, curcumin, resveratrol, and silymarin are the most extensively studied; however, their differential effects on MASLD outcomes remain inconclusive. This systematic review and meta-analysis of RCTs aimed to evaluate the efficacy of curcumin, resveratrol, and silymarin in patients with MASLD. A comprehensive search of seven databases was conducted up to September 2024. Odds ratios (OR), mean differences (MD), and standardized MD (SMD) with 95% confidence intervals (CI) were used to assess treatment effects. Primary outcomes included improvement in hepatic steatosis and ALT activity, while secondary outcomes included changes in AST activity, blood lipids, glucose, BMI, blood pressure, and TNF-α. Twenty-seven studies involving 1691 participants were included. Curcumin significantly improved hepatic steatosis compared to placebo (OR: 4.39, 95% CI: 1.45 to 13.27, p = 0.009), followed by resveratrol (OR: 3.18, 95% CI: 1.20 to 8.42, p = 0.02). Silymarin exhibited the strongest effect in reducing ALT levels (MD: -6.44 U/L, 95% CI: -10.03 to -2.85, p = 0.0004), with curcumin (MD: -5.88 U/L, 95% CI: -9.05 to -2.72, p = 0.0003) also showing significant reductions. A marked reduction in AST was observed with silymarin (MD: -6.99 U/L, 95% CI: -8.56 to -5.42, p < 0.00001), followed by curcumin (MD: -3.36 U/L, 95% CI: -5.35 to -1.36, p = 0.001). Furthermore, curcumin intake significantly improved metabolic indicators (TG, FBG, HOMA-IR, and BMI). Resveratrol reduced FBG and DBP. Curcumin had the strongest effect on hepatic steatosis and improved both transaminase levels and metabolic markers. Silymarin demonstrated the greatest reduction in transaminase levels, while resveratrol showed modest benefits in steatosis and metabolic improvements. The three polyphenols appear as promising therapeutics for the treatment of MASLD.
Collapse
Affiliation(s)
- Qian Huang
- Institute of Liver DiseasesShuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
- School of Basic MedicineShaanxi University of Chinese MedicineXianyangShaanxiChina
| | - Ziming An
- Institute of Liver DiseasesShuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Xin Xin
- Institute of Liver DiseasesShuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Xiaojun Gou
- Central LaboratoryBaoshan District Hospital of Integrated Traditional Chinese and Western Medicine of ShanghaiShanghaiChina
| | - Xiaoting Tian
- Shanghai Institute of Materia Medica, Chinese Academy of SciencesShanghaiChina
| | - Yiyang Hu
- Institute of Liver DiseasesShuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Zubing Mei
- Institute of Anorectal DiseasesShuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Qin Feng
- Institute of Liver DiseasesShuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
- Shanghai Key Laboratory of Traditional Chinese Clinical MedicineShanghaiChina
- Key Laboratory of Liver and Kidney DiseasesShanghai University of Traditional Chinese Medicine, Ministry of EducationShanghaiChina
- Central LaboratoryShuguang Hospital Affiliated to Shanghai University of Chinese Traditional MedicineShanghaiChina
| |
Collapse
|
5
|
Asadi A, Rostami M, Shafiee R, Ardalani A, Dehghanitafti A, Golshadi Z, Kohansal K, Ghasemi F, Najafi M, Mahmoudi T, Rezamand G, Dabiri R, Nobakht H, Farahani H, Tabaeian SP. Association of IRS1 gene Pro512Ala polymorphism with nonalcoholic fatty liver disease. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2024; 68:e230216. [PMID: 39420901 PMCID: PMC11460970 DOI: 10.20945/2359-4292-2023-0216] [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: 09/13/2023] [Accepted: 10/02/2023] [Indexed: 10/19/2024]
Abstract
Objective This study was designed to investigate the possible effect of the insulin receptor substrate 1 (IRS1) gene rs1801276 polymorphism on the risk of nonalcoholic fatty liver disease (NAFLD). Subjects and methods The rs1801276 polymorphism was investigated in 127 controls and 123 biopsy-proven NAFLD patients using PCR-RFLP. Results No deviation from Hardy-Weinberg equilibrium was discovered for the rs1801276 variant of IRS1 in either NAFLD patients or controls (P>0.05). The distribution of different rs1801276 genotypes and alleles showed significant variations between controls and NAFLD patients. In comparison to rs1801276 'CC' genotype, the "GG+GC" genotype occurred less frequently in NAFLD patients than in controls, which also persisted after adjustment for confounding factors (P = 0.041, OR = 0.60, 95% CI = 0.45-0.93). In comparison with the IRS1 rs1801276 "C" allele, the "G" allele was significantly less prevalent in NAFLD patients than in controls (P = 0.045, OR = 0.69, 95% CI = 0.58-0.91). Conclusions For the first time, we reported a significant association between the IRS1 rs1801276 polymorphism and biopsy-proven NAFLD. More studies are required to further elucidate the contribution of the IRS1 gene to NAFLD susceptibility.
Collapse
Affiliation(s)
- Asadollah Asadi
- University of Mohaghegh ArdabiliFaculty of ScienceDepartment of BiologyArdabilIranDepartment of Biology, Faculty of Science, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Mitra Rostami
- Shahid Beheshti University of Medical SciencesResearch Institute for Gastroenterology and Liver DiseasesGastroenterology and Liver Diseases Research CenterTehranIranGastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Radmehr Shafiee
- Tehran UniversityFaculty of Veterinary MedicineDepartment of Clinical PathologyIranDepartment of Clinical Pathology, Faculty of Veterinary Medicine, Tehran University, Iran
| | - Abbas Ardalani
- Shahid Beheshti University of Medical SciencesResearch Institute for Gastroenterology and Liver DiseasesGastroenterology and Liver Diseases Research CenterTehranIranGastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atefeh Dehghanitafti
- Shahid Beheshti University of Medical SciencesResearch Institute for Gastroenterology and Liver DiseasesGastroenterology and Liver Diseases Research CenterTehranIranGastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zakieh Golshadi
- Shahid Beheshti University of Medical SciencesResearch Institute for Gastroenterology and Liver DiseasesGastroenterology and Liver Diseases Research CenterTehranIranGastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kiarash Kohansal
- Iran University of Medical SciencesPhysiology Research CenterTehranIranPhysiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ghasemi
- Shahid Beheshti University of Medical SciencesResearch Institute for Gastroenterology and Liver DiseasesGastroenterology and Liver Diseases Research CenterTehranIranGastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Najafi
- Shahid Beheshti University of Medical SciencesResearch Institute for Gastroenterology and Liver DiseasesGastroenterology and Liver Diseases Research CenterTehranIranGastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Touraj Mahmoudi
- Shahid Beheshti University of Medical SciencesResearch Institute for Gastroenterology and Liver DiseasesGastroenterology and Liver Diseases Research CenterTehranIranGastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Gholamreza Rezamand
- Iran University of Medical SciencesSchool of MedicineDepartment of Internal MedicineTehranIranDepartment of Internal Medicine, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Iran University of Medical SciencesColorectal Research CenterTehranIranColorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Dabiri
- Semnan University of Medical SciencesInternal Medicine DepartmentSemnanIranInternal Medicine Department, Semnan University of Medical Sciences, Semnan, Iran
| | - Hossein Nobakht
- Semnan University of Medical SciencesInternal Medicine DepartmentSemnanIranInternal Medicine Department, Semnan University of Medical Sciences, Semnan, Iran
| | - Hamid Farahani
- Qom University of Medical SciencesSchool of MedicineDepartment of Physiology and PharmacologyQomIranDepartment of Physiology and Pharmacology, School of Medicine, Qom University of Medical Sciences, Qom, Iran
| | - Seidamir Pasha Tabaeian
- Iran University of Medical SciencesSchool of MedicineDepartment of Internal MedicineTehranIranDepartment of Internal Medicine, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Iran University of Medical SciencesColorectal Research CenterTehranIranColorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Zhang X, Liu M, Wang Z, Wang P, Kong L, Wu J, Wu W, Ma L, Jiang S, Ren W, Du L, Ma W, Liu X. A review of the botany, phytochemistry, pharmacology, synthetic biology and comprehensive utilization of Silybum marianum. Front Pharmacol 2024; 15:1417655. [PMID: 39055491 PMCID: PMC11269164 DOI: 10.3389/fphar.2024.1417655] [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: 04/15/2024] [Accepted: 06/20/2024] [Indexed: 07/27/2024] Open
Abstract
Silybum marianum (L.) Gaertn, a herbaceous plant with a long history in traditional medicine for the treatment of hepatobiliary diseases, particularly in Europe, which has attracted attention for its remarkable therapeutic effect. This review systematically summarizes the research progress in the botany, phytochemistry, pharmacology, comprehensive utilization and synthetic biology of S. marianum. Up to now, more than 20 types of flavonolignan components have been isolated from S. marianum. In addition, the rearch on fatty acids and triterpenoids is also constantly improving. Among them, silybin is the most active compound in flavonolignans components. Its pharmacological effects in vivo and in vitro include anti-inflammatory, antioxidant, anti-tumour, hypoglycaemic, neuroprotective and immunoregulatory properties. The use of coniferyl alcohol and taxifolin as substrates to produce silybin and isosilybin under the action of enzyme catalysis is the commonly used biosynthetic pathway of silymarin, which provides support for a comprehensive analysis of the synthetic pathway of silymarin. In addition to medicinal use, the extracts of plants also have broad application prospects in the production of food, healthcare products, cosmetics and other aspects. In addition, the chemical composition, pharmacological mechanism and synthetic biology of S. marianum need to be further studied, which is very important for its clinical efficacy and resource development.
Collapse
Affiliation(s)
- Xiaozhuang Zhang
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Meiqi Liu
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zhen Wang
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Panpan Wang
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Lingyang Kong
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jianhao Wu
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Wei Wu
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Lengleng Ma
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Shan Jiang
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Weichao Ren
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Likun Du
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Wei Ma
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xiubo Liu
- College of Jiamusi, Heilongjiang University of Chinese Medicine, Jiamusi, China
| |
Collapse
|
7
|
MacDonald-Ramos K, Monroy A, Bobadilla-Bravo M, Cerbón M. Silymarin Reduced Insulin Resistance in Non-Diabetic Women with Obesity. Int J Mol Sci 2024; 25:2050. [PMID: 38396727 PMCID: PMC10888588 DOI: 10.3390/ijms25042050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 01/25/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
Silymarin has ameliorated obesity, type 2 diabetes (T2DM), and insulin resistance (IR) in combination with standard therapy, diet, or exercise in recent studies. Obesity and IR are the main risk factors for developing T2DM and other metabolic disorders. Today, there is a need for new strategies to target IR in patients with these metabolic diseases. In the present longitudinal study, a group of non-diabetic insulin-resistant women with type 1 and type 2 obesity were given silymarin for 12 weeks, with no change in habitual diet and physical activity. We used the Homeostatic Model Assessment for Insulin Resistance Index (HOMA-IR) to determine IR at baseline and after silymarin treatment (t = 12 weeks). We obtained five timepoint oral glucose tolerance tests, and other biochemical and clinical parameters were analyzed before and after treatment. Treatment with silymarin alone significantly reduced mean fasting plasma glucose (FPG) and HOMA-IR levels at 12 weeks compared to baseline values (p < 0.05). Mean fasting plasma insulin (FPI), total cholesterol, high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), triglycerides (Tg), indirect bilirubin, and C-reactive protein (CRP) levels decreased compared to baseline values, although changes were non-significant. The overall results suggest that silymarin may offer a therapeutic alternative to improve IR in non-diabetic individuals with obesity. Further clinical trials are needed in this type of patient to strengthen the results of this study.
Collapse
Affiliation(s)
- Karla MacDonald-Ramos
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
- Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| | - Adriana Monroy
- Servicio de Oncología, Hospital General de México Dr. Eduardo Liceaga, Ciudad de México 06720, Mexico;
| | - Mariana Bobadilla-Bravo
- Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| | - Marco Cerbón
- Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| |
Collapse
|
8
|
Chen X, Hou Y, Liao A, Pan L, Yang S, Liu Y, Wang J, Xue Y, Zhang M, Zhu Z, Huang J. Integrated Analysis of Gut Microbiome and Adipose Transcriptome Reveals Beneficial Effects of Resistant Dextrin from Wheat Starch on Insulin Resistance in Kunming Mice. Biomolecules 2024; 14:186. [PMID: 38397423 PMCID: PMC10886926 DOI: 10.3390/biom14020186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 01/14/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
Systemic chronic inflammation is recognized as a significant contributor to the development of obesity-related insulin resistance. Previous studies have revealed the physiological benefits of resistant dextrin (RD), including obesity reduction, lower fasting glucose levels, and anti-inflammation. The present study investigated the effects of RD intervention on insulin resistance (IR) in Kunming mice, expounding the mechanisms through the gut microbiome and transcriptome of white adipose. In this eight-week study, we investigated changes in tissue weight, glucose-lipid metabolism levels, serum inflammation levels, and lesions of epididymal white adipose tissue (eWAT) evaluated via Hematoxylin and Eosin (H&E) staining. Moreover, we analyzed the gut microbiota composition and transcriptome of eWAT to assess the potential protective effects of RD intervention. Compared with a high-fat, high-sugar diet (HFHSD) group, the RD intervention significantly enhanced glucose homeostasis (e.g., AUC-OGTT, HOMA-IR, p < 0.001), and reduced lipid metabolism (e.g., TG, LDL-C, p < 0.001) and serum inflammation levels (e.g., IL-1β, IL-6, p < 0.001). The RD intervention also led to changes in the gut microbiota composition, with an increase in the abundance of probiotics (e.g., Parabacteroides, Faecalibaculum, and Muribaculum, p < 0.05) and a decrease in harmful bacteria (Colidextribacter, p < 0.05). Moreover, the RD intervention had a noticeable effect on the gene transcription profile of eWAT, and KEGG enrichment analysis revealed that differential genes were enriched in PI3K/AKT, AMPK, in glucose-lipid metabolism, and in the regulation of lipolysis in adipocytes signaling pathways. The findings demonstrated that RD not only ameliorated IR, but also remodeled the gut microbiota and modified the transcriptome profile of eWAT.
Collapse
Affiliation(s)
- Xinyang Chen
- Food Laboratory of Zhongyuan, Luohe 462300, China; (X.C.); (Y.H.); (A.L.); (L.P.); (Y.L.); (J.W.); (Y.X.); (M.Z.); (Z.Z.)
- School of Biological Engineering, Henan University of Technology, Zhengzhou 450001, China
| | - Yinchen Hou
- Food Laboratory of Zhongyuan, Luohe 462300, China; (X.C.); (Y.H.); (A.L.); (L.P.); (Y.L.); (J.W.); (Y.X.); (M.Z.); (Z.Z.)
- College of Food and Biological Engineering, Henan University of Animal Husbandry and Economy, Zhengzhou 450046, China;
| | - Aimei Liao
- Food Laboratory of Zhongyuan, Luohe 462300, China; (X.C.); (Y.H.); (A.L.); (L.P.); (Y.L.); (J.W.); (Y.X.); (M.Z.); (Z.Z.)
- School of Biological Engineering, Henan University of Technology, Zhengzhou 450001, China
| | - Long Pan
- Food Laboratory of Zhongyuan, Luohe 462300, China; (X.C.); (Y.H.); (A.L.); (L.P.); (Y.L.); (J.W.); (Y.X.); (M.Z.); (Z.Z.)
- School of Biological Engineering, Henan University of Technology, Zhengzhou 450001, China
| | - Shengru Yang
- College of Food and Biological Engineering, Henan University of Animal Husbandry and Economy, Zhengzhou 450046, China;
| | - Yingying Liu
- Food Laboratory of Zhongyuan, Luohe 462300, China; (X.C.); (Y.H.); (A.L.); (L.P.); (Y.L.); (J.W.); (Y.X.); (M.Z.); (Z.Z.)
- School of Biological Engineering, Henan University of Technology, Zhengzhou 450001, China
| | - Jingjing Wang
- Food Laboratory of Zhongyuan, Luohe 462300, China; (X.C.); (Y.H.); (A.L.); (L.P.); (Y.L.); (J.W.); (Y.X.); (M.Z.); (Z.Z.)
- School of Biological Engineering, Henan University of Technology, Zhengzhou 450001, China
| | - Yingchun Xue
- Food Laboratory of Zhongyuan, Luohe 462300, China; (X.C.); (Y.H.); (A.L.); (L.P.); (Y.L.); (J.W.); (Y.X.); (M.Z.); (Z.Z.)
- School of Biological Engineering, Henan University of Technology, Zhengzhou 450001, China
| | - Mingyi Zhang
- Food Laboratory of Zhongyuan, Luohe 462300, China; (X.C.); (Y.H.); (A.L.); (L.P.); (Y.L.); (J.W.); (Y.X.); (M.Z.); (Z.Z.)
- School of Biological Engineering, Henan University of Technology, Zhengzhou 450001, China
| | - Zhitong Zhu
- Food Laboratory of Zhongyuan, Luohe 462300, China; (X.C.); (Y.H.); (A.L.); (L.P.); (Y.L.); (J.W.); (Y.X.); (M.Z.); (Z.Z.)
- School of Biological Engineering, Henan University of Technology, Zhengzhou 450001, China
| | - Jihong Huang
- Food Laboratory of Zhongyuan, Luohe 462300, China; (X.C.); (Y.H.); (A.L.); (L.P.); (Y.L.); (J.W.); (Y.X.); (M.Z.); (Z.Z.)
- School of Biological Engineering, Henan University of Technology, Zhengzhou 450001, China
- State Key Laboratory of Crop Stress Adaptation and Improvement, College of Agriculture, Henan University, Kaifeng 475004, China
- School of Food and Pharmacy, Xuchang University, Xuchang 461000, China
| |
Collapse
|
9
|
Ma X, Yu X, Li R, Cui J, Yu H, Ren L, Jiang J, Zhang W, Wang L. Berberine-silybin salt achieves improved anti-nonalcoholic fatty liver disease effect through regulating lipid metabolism. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117238. [PMID: 37774895 DOI: 10.1016/j.jep.2023.117238] [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: 06/26/2023] [Revised: 08/15/2023] [Accepted: 09/26/2023] [Indexed: 10/01/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Berberine (BBR) and silybin (SIY) are natural compounds obtained from Berberidaceae members and Silybum marianum (L.) Gaertn., respectively. These compounds have been demonstrated to regulate lipid metabolism and indue hepatoprotective effects, establishing their importance for the treatment of liver injury. Combination therapy has shown promise in treating ailments with complex pathophysiology, such as liver diseases. However, the inconsistent dissolution and poor absorption of BBR and SIY limit their efficacy. AIM OF THE STUDY This study compared the salt formulation (BSS) and physical mixture (BSP) of BBR and SIY for their efficacy in treating nonalcoholic fatty liver disease (NAFLD). MATERIALS AND METHODS The formation of the BSS was confirmed using various techniques, including nuclear magnetic resonance spectroscopy, Fourier-transform infrared spectroscopy, differential scanning calorimetry, scanning electron microscopy, and powder X-ray diffractometry. In addition, dissolution, trans-epithelial permeability, and bioavailability experiments were conducted to evaluate the absorption and distribution of drugs. Pharmacodynamics and mechanisms were investigated through in vivo experiments. RESULTS BSS form demonstrated synchronized dissolution of both components, unlike BSP. Additionally, the transepithelial permeability results revealed that BSS exhibited superior penetration and absorption of both BBR and SIY in comparison to BSP. Furthermore, BSS significantly increased the bioavailability of SIY in both plasma and the liver (2.2- and 4.5-fold, respectively) when compared with BSP. Moreover, BSS demonstrated a more potent inhibitory effect on lipid production in HepG2 cells than BSP. In mouse models (BALB/c) of NAFLD, BSS improved disease outcomes, as evidenced by decreased adipose levels, normalized blood lipid levels, and reduced liver parenchyma injury. Preliminary transcriptomics analysis suggested that BSS achieved its anti-NAFLD effect by regulating the expression of fatty acid transporter CD36, recombinant fatty acid binding protein 4, and stearyl coenzyme A dehydrogenase 1, which are associated with the synthesis and uptake of fatty acid-related proteins. CONCLUSIONS The study demonstrated that compared with physical mixing, salification improved the efficacy of BBR and SIY, as demonstrated in animal experiments. These findings provide valuable insights into the development of more effective treatments for NAFLD and provide new possibilities for combination therapies.
Collapse
Affiliation(s)
- Xiaolei Ma
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xiaoyou Yu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Rui Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Jinjin Cui
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Haoyang Yu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Ling Ren
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Jiandong Jiang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China; Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.
| | - Wenxuan Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.
| | - Lulu Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.
| |
Collapse
|
10
|
Di Sarli Gutiérrez L, Castro MC, Farromeque Vásquez S, Villagarcía HG, González Arbeláez L, Rojano B, Schinella G, Maiztegui B, Francini F. Protective Effect of Monoterpene Isoespintanol in a Rat Model of Prediabetes Induced by Fructose. Pharmaceuticals (Basel) 2023; 17:47. [PMID: 38256882 PMCID: PMC10819293 DOI: 10.3390/ph17010047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 09/21/2023] [Accepted: 09/25/2023] [Indexed: 01/24/2024] Open
Abstract
A high-fructose diet (HFD) induces murine alterations like those recorded in human prediabetes. Protective effects of isoespintanol (monoterpene isolated from Oxandra cf. xylopioides) on changes induced by HFD were evaluated. Animals were maintained for 21 days with a standard diet (C), 10% fructose (F), and F plus isoespintanol (FI, 10 mg/kg, i.p.). Glycemia, triglyceridemia, total and HDL-cholesterol, and insulin resistance index (IRX) were determined. Intraperitoneal glucose tolerance test (IGTT) was performed. In the liver, we measured glycogen, lipogenic gene expression (SREBP-1c, GPAT, FAS, and CPT1), oxidative stress (GSH and 3'-nitrotyrosine content), inflammation markers (iNOS, TNF-α, and PAI-1 gene expression; iNOS and COX-2 protein levels), p-eNOS, p-Akt, and p-GSK3β protein levels. Isoespintanol corrected enhanced triglycerides, lipogenic genes, and IRX, and reduced HDL-cholesterol induced by HFD. Increased liver glycogen and inflammatory markers and decreased GSH, p-Akt, and p-GSK3β measured in F rats were reversed by isoespintanol, and p-eNOS/e-NOS and iNOS/GADPH ratios were normalized. Isoespintanol restored glucose tolerance (IGTT) compared to F rats. These results demonstrate for the first time that isoespintanol prevents endocrine-metabolic alterations induced by HFD in prediabetic rats. These effects could be mediated by Akt/eNOS and Akt/GSK3β pathways, suggesting its possible use as a therapeutic tool for the prevention of diabetes at early stages of its development (prediabetes).
Collapse
Affiliation(s)
- Luciana Di Sarli Gutiérrez
- CENEXA (Centre for Experimental and Applied Endocrinology, UNLP CONICET CCT La Plata, CEAS CICPBA), School of Medicine, Street 60 and 120, La Plata 1900, Argentina; (L.D.S.G.); (M.C.C.); (S.F.V.); (H.G.V.); (B.M.)
| | - María Cecilia Castro
- CENEXA (Centre for Experimental and Applied Endocrinology, UNLP CONICET CCT La Plata, CEAS CICPBA), School of Medicine, Street 60 and 120, La Plata 1900, Argentina; (L.D.S.G.); (M.C.C.); (S.F.V.); (H.G.V.); (B.M.)
| | - Sherley Farromeque Vásquez
- CENEXA (Centre for Experimental and Applied Endocrinology, UNLP CONICET CCT La Plata, CEAS CICPBA), School of Medicine, Street 60 and 120, La Plata 1900, Argentina; (L.D.S.G.); (M.C.C.); (S.F.V.); (H.G.V.); (B.M.)
| | - Hernán Gonzalo Villagarcía
- CENEXA (Centre for Experimental and Applied Endocrinology, UNLP CONICET CCT La Plata, CEAS CICPBA), School of Medicine, Street 60 and 120, La Plata 1900, Argentina; (L.D.S.G.); (M.C.C.); (S.F.V.); (H.G.V.); (B.M.)
| | - Luisa González Arbeláez
- CIC (Centre for Cardiovascular Research, UNLP CONICET CCT La Plata), School of Medicine, Street 60 and 120, La Plata 1900, Argentina;
| | - Benjamín Rojano
- Food Science Laboratory, Faculty of Sciences, National University of Colombia, Medellín Campus, Medellin 050034, Colombia;
| | - Guillermo Schinella
- School of Medicine, UNLP, Street 60 and 120, La Plata 1900, Argentina;
- Institute of Health Sciences, UNAJ-CICPBA, Av. Calchaquí 6200, Florencio Varela 1888, Argentina
| | - Bárbara Maiztegui
- CENEXA (Centre for Experimental and Applied Endocrinology, UNLP CONICET CCT La Plata, CEAS CICPBA), School of Medicine, Street 60 and 120, La Plata 1900, Argentina; (L.D.S.G.); (M.C.C.); (S.F.V.); (H.G.V.); (B.M.)
| | - Flavio Francini
- CENEXA (Centre for Experimental and Applied Endocrinology, UNLP CONICET CCT La Plata, CEAS CICPBA), School of Medicine, Street 60 and 120, La Plata 1900, Argentina; (L.D.S.G.); (M.C.C.); (S.F.V.); (H.G.V.); (B.M.)
| |
Collapse
|
11
|
Wang W, Zhai T, Luo P, Miao X, Wang J, Chen Y. Beneficial effects of silibinin on serum lipids, bile acids, and gut microbiota in methionine-choline-deficient diet-induced mice. Front Nutr 2023; 10:1257158. [PMID: 37867498 PMCID: PMC10587424 DOI: 10.3389/fnut.2023.1257158] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 09/11/2023] [Indexed: 10/24/2023] Open
Abstract
Background and purpose Silibinin (SIL) is a flavonoid lignin isolated from the fruit and seeds of silybum marianum that exhibits good therapeutic potential for NASH. However, the effects of SIL on serum lipids, bile acids (BAs), and gut microbiota (GM) in NASH mice remain unknown. The present work aimed to explore the beneficial effects of SIL supplementation on serum lipids, bile acids, and gut microbiota in MCD mice. Experimental approach After male C57BL/6 mice were fed with a methionine-choline deficient (MCD) diet and simultaneously gavaged with SIL (20 mg/kg. d) for 8 weeks, the pathological changes of liver tissue were observed by oil red O, haematoxylin-eosin, and Masson tricolor staining; the levels of serum AST and ALT, and liver TG and MDA were detected by assay kits; metabonomics and 16S rDNA sequencing were used to analyze the composition of serum lipids and BAs and the abundance of GM; and the mRNA expression levels of hepatic genes related to BAs homeostasis were detected by RT-qPCR. Results The results indicated that SIL treatment decreased the levels of 26 lipids (including four arachidonic acids, seven FFAs, 12 acyl carnitines, and three GPs) and two BAs (23-DCA, GLCA), while Dubosiella increased the levels of 10 lipids (including TxB3, PG16:0_18:1, Cer t18:0/24:0 and 7 TGs), five BAs (β-MCA, α-MCA, UDCA, 3-oxo-DCA and HCA), and two GMs (Verrucomicrobiota and Akkermansiaceae) of MCD mice, but had no significant effect on the mRNA expression of CYP7A1, CYP27A1, Bsep, Mrp2, Ntcp, or Oatp1b2. Therefore, influencing GM composition and then regulating the levels of serum lipids and BAs through enterohepatic axis should be an important mechanism of SIL-induced alleviative effect on MCD mice. More importantly, we found that SIL had a good coordination in regulating the abundance of GM and the contents of serum lipids and BAs in MCD mice, that is, when the abundance of probiotics was up-regulated, the content of beneficial unsaturated fatty acids in serum was up-regulated, while the serum levels of harmful lipids and BAs were down-regulated. Conclusion The alleviating effect of SIL on NASH may be closely related to the correction of intestinal bacteria disorder, serum bile acid, and lipid metabolic disturbance in mice.
Collapse
Affiliation(s)
- Wei Wang
- Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, National and Local Joint Engineering Research Center of High-throughput Drug Screening Technology, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, China
| | - Ting Zhai
- Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, National and Local Joint Engineering Research Center of High-throughput Drug Screening Technology, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, China
| | - Ping Luo
- School of Pharmacy, Hubei University of Science and Technology, Xianning, China
| | - Xiaolei Miao
- School of Pharmacy, Hubei University of Science and Technology, Xianning, China
| | - Junjun Wang
- Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, National and Local Joint Engineering Research Center of High-throughput Drug Screening Technology, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, China
| | - Yong Chen
- Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, National and Local Joint Engineering Research Center of High-throughput Drug Screening Technology, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, China
| |
Collapse
|
12
|
Yuan P, Zhang Q, Fu Y, Hou Y, Gao L, Wei Y, Feng W, Zheng X. Acacetin inhibits myocardial mitochondrial dysfunction by activating PI3K/AKT in SHR rats fed with fructose. J Nat Med 2023; 77:262-275. [PMID: 36520340 DOI: 10.1007/s11418-022-01666-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 11/23/2022] [Indexed: 12/23/2022]
Abstract
To explore the effect of acacetin on myocardial mitochondrial dysfunction in spontaneously hypertensive rats (SHR) with insulin resistance (IR), and the possible mechanism. Rapid IR was first induced in fructose-fed SHR, and they were then treated with acacetin (25, 50 mg/kg). After 7 weeks, the rats were tested for hypertension, IR, cardiac function, and mitochondrial damage status. Potential mechanisms of action were explored in terms of oxidative stress, mitochondrial fission and division, apoptosis, and the insulin signaling pathway. Subsequently, the PI3K gene was silenced, after intervention with acacetin (5 μM) for 24 h, and H2O2 was used to stimulate H9c2 for 4 h, it was evaluated whether silencing PI3K would affect the therapeutic effect of acacetin. In SHR fed with fructose, acacetin can improve hypertension, IR, cardiac function (LVEF, LVFS), and mitochondrial damage (mitochondria number, ATP); inhibit oxidative stress (ROS, SOD, Nrf2, Keap1), mitochondrial fission (MFF, Drp1), and myocardial cell apoptosis (apoptosis rate, Bax, Bcl-2, cytochrome c); promote mitochondrial fusion (Mfn2) and activate insulin signaling pathways (PI3K/AKT). However, silencing PI3K inhibited the abovementioned effects of acacetin. In conclusion, acacetin improved myocardial mitochondrial dysfunction through regulating oxidative stress, mitochondrial fission and fusion, and mitochondrial pathway apoptosis mediated by PI3K/AKT signaling pathway in hypertensive rats with IR.
Collapse
Affiliation(s)
- Peipei Yuan
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, 450046, China
| | - Qi Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Yang Fu
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Ying Hou
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Liyuan Gao
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Yaxin Wei
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Weisheng Feng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, 450046, China
| | - Xiaoke Zheng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, 450046, China.
| |
Collapse
|
13
|
Jeeyavudeen MS, Khan SKA, Fouda S, Pappachan JM. Management of metabolic-associated fatty liver disease: The diabetology perspective. World J Gastroenterol 2023; 29:126-143. [PMID: 36683717 PMCID: PMC9850951 DOI: 10.3748/wjg.v29.i1.126] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/26/2022] [Accepted: 12/13/2022] [Indexed: 01/04/2023] Open
Abstract
The metabolic syndrome as a consequence of the obesity pandemic resulted in a substantial increase in the prevalence of metabolic-associated fatty live disease (MAFLD) and type 2 diabetes mellitus (T2DM). Because of the similarity in pathobiology shared between T2DM and MAFLD, both disorders coexist in many patients and may potentiate the disease-related outcomes with rapid progression and increased complications of the individual diseases. In fact, awareness about this coexistence and the risk of complications are often overlooked by both hepatologists and diabetologists. Management of these individual disorders in a patient should be addressed wholistically using an appropriate multidisciplinary team approach involving both the specialists and, when necessary, liaising with dieticians and surgeons. This comprehensive review is to compile the current evidence from a diabetologist's perspective on MAFLD and T2DM and to suggest optimal management strategies.
Collapse
Affiliation(s)
- Mohammad Sadiq Jeeyavudeen
- Department of Endocrinology and Metabolism, University Hospitals of Edinburgh, Edinburgh EH4 2XU, United Kingdom
| | - Shahanas K A Khan
- Department of Endocrinology and Metabolism, Lancashire Teaching Hospitals NHS Trust, Preston PR2 9HT, United Kingdom
| | - Sherouk Fouda
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC 3046, Australia
| | - Joseph M Pappachan
- Department of Endocrinology and Metabolism, Lancashire Teaching Hospitals NHS Trust, Preston PR2 9HT, United Kingdom
- Faculty of Science, Manchester Metropolitan University, Manchester M15 6BH, United Kingdom
- Faculty of Biology, University of Manchester, Manchester M13 9PL, United Kingdom
| |
Collapse
|
14
|
Dong M, Chen H, Wen S, Yuan Y, Yang L, Li Y, Yuan X, Xu D, Zhou L. The Neuronal and Non-Neuronal Pathways of Sodium-Glucose Cotransporter-2 Inhibitor on Body Weight-Loss and Insulin Resistance. Diabetes Metab Syndr Obes 2023; 16:425-435. [PMID: 36820270 PMCID: PMC9938665 DOI: 10.2147/dmso.s399367] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
With the emergence of sodium-glucose cotransporter 2 inhibitors (SGLT2i), the treatment of type 2 diabetes mellitus (T2DM) has achieved a new milestone, of which the insulin-independent mechanism could produce weight loss, improve insulin resistance (IR) and exert other protective effects. Besides the well-acknowledged biochemical processes, the dysregulated balance between sympathetic and parasympathetic activity may play a significant role in IR and obesity. Weight loss caused by SGLT-2i could be achieved via activating the liver-brain-adipose neural axis in adipocytes. We previously demonstrated that SGLT-2 are widely expressed in central nervous system (CNS) tissues, and SGLT-2i could inhibit central areas associated with autonomic control through unidentified pathways, indicating that the role of the central sympathetic inhibition of SGLT-2i on blood pressure and weight loss. However, the exact pathway of SGLT2i related to these effects and to what extent it depends on the neural system are not fully understood. The evidence of how SGLT-2i interacts with the nervous system is worth exploring. Therefore, in this review, we will illustrate the potential neurological processes by which SGLT2i improves IR in skeletal muscle, liver, adipose tissue, and other insulin-target organs via the CNS and sympathetic nervous system/parasympathetic nervous system (SNS/PNS).
Collapse
Affiliation(s)
- Meiyuan Dong
- Graduate School of Hebei Medical University, Shijiazhuang, People’s Republic of China
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Huiling Chen
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Song Wen
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Yue Yuan
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Liling Yang
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Yanyan Li
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Xinlu Yuan
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Dongxiang Xu
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Ligang Zhou
- Graduate School of Hebei Medical University, Shijiazhuang, People’s Republic of China
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling, Shanghai Pudong Hospital, Shanghai, People’s Republic of China
- Correspondence: Ligang Zhou, Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, People’s Republic of China, Tel +8613611927616, Email
| |
Collapse
|
15
|
Upregulation of Klotho Aggravates Insulin Resistance in Gestational Diabetes Mellitus Trophoblast Cells. Genet Res (Camb) 2022; 2022:1500768. [DOI: 10.1155/2022/1500768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 09/07/2022] [Accepted: 09/15/2022] [Indexed: 11/17/2022] Open
Abstract
Objective. Insulin resistance (IR) plays a key role in gestational diabetes mellitus (GDM) pathogenesis. The antiaging protein klotho has been proven to be closely related to IR. The purpose of this study was to investigate the effect of klotho on IR in GDM trophoblast cells. Methods. The GDM cell model of HTR-8/SVneo cells was induced by high glucose (HG). Plasmid transfection was used to mediate the overexpression or silencing of klotho. The effects of klotho on cell viability, IR, and the IGF-1/PI3K pathways were observed by RT-qPCR, western blot, Cell Counting Kit-8 detection, glucose uptake assay, and immunofluorescence detection. Results. Klotho expression was up-regulated in HG-induced cells. Overexpression of klotho could reduce the cell viability, insulin signaling molecules (INSR-α, INSR-β, IRS1, IRS2, and GLUT4), and glucose uptake in HTR-8/SVneo cells of the HG group. In addition, the overexpression of klotho inhibited the levels of IGF-1, IGF-1R/p-IGF-1R, and the phosphorylation and activation of the signal transduction molecules PI3K/Akt/mTOR. On the contrary, klotho deletions could reverse these changes of HTR-8/SVneo cells induced by HG. Conclusion. In a word, the results of this study showed that the regulation of klotho played an important role in the IR of trophoblast cells induced by HG, which was mediated at least in part by the IGF-1/PI3K/Akt/mTOR pathway.
Collapse
|
16
|
Effect of a hexacyclic triterpenic acid from Euscaphis japonica on the oleic acid induced HepG2 cellular model of non-alcoholic fatty liver disease. Med Chem Res 2022. [DOI: 10.1007/s00044-022-02982-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
17
|
Shao G, Liu Y, Lu L, Zhang G, Zhou W, Wu T, Wang L, Xu H, Ji G. The Pathogenesis of HCC Driven by NASH and the Preventive and Therapeutic Effects of Natural Products. Front Pharmacol 2022; 13:944088. [PMID: 35873545 PMCID: PMC9301043 DOI: 10.3389/fphar.2022.944088] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 06/20/2022] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is a clinical syndrome with pathological changes that are similar to those of alcoholic hepatitis without a history of excessive alcohol consumption. It is a specific form of nonalcoholic fatty liver disease (NAFLD) that is characterized by hepatocyte inflammation based on hepatocellular steatosis. Further exacerbation of NASH can lead to cirrhosis, which may then progress to hepatocellular carcinoma (HCC). There is a lack of specific and effective treatments for NASH and NASH-driven HCC, and the mechanisms of the progression of NASH to HCC are unclear. Therefore, there is a need to understand the pathogenesis and progression of these diseases to identify new therapeutic approaches. Currently, an increasing number of studies are focusing on the utility of natural products in NASH, which is likely to be a promising prospect for NASH. This paper reviews the possible mechanisms of the pathogenesis and progression of NASH and NASH-derived HCC, as well as the potential therapeutic role of natural products in NASH and NASH-derived HCC.
Collapse
Affiliation(s)
- Gaoxuan Shao
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ying Liu
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lu Lu
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guangtao Zhang
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenjun Zhou
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tao Wu
- Institute of Interdisciplinary Integrative Biomedical Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lei Wang
- Department of Hepatology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hanchen Xu
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guang Ji
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
18
|
Lee HA, Chang Y, Sung PS, Yoon EL, Lee HW, Yoo JJ, Lee YS, An J, Song DS, Cho YY, Kim SU, Kim YJ. Therapeutic mechanisms and beneficial effects of non-antidiabetic drugs in chronic liver diseases. Clin Mol Hepatol 2022; 28:425-472. [PMID: 35850495 PMCID: PMC9293616 DOI: 10.3350/cmh.2022.0186] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 06/29/2022] [Indexed: 11/05/2022] Open
Abstract
The global burden of chronic liver disease (CLD) is substantial. Due to the limited indication of and accessibility to antiviral therapy in viral hepatitis and lack of effective pharmacological treatment in nonalcoholic fatty liver disease, the beneficial effects of antidiabetics and non-antidiabetics in clinical practice have been continuously investigated in patients with CLD. In this narrative review, we focused on non-antidiabetic drugs, including ursodeoxycholic acid, silymarin, dimethyl4,4'-dimethoxy-5,6,5',6'-dimethylenedixoybiphenyl-2,2'-dicarboxylate, L-ornithine L-aspartate, branched chain amino acids, statin, probiotics, vitamin E, and aspirin, and summarized their beneficial effects in CLD. Based on the antioxidant, anti-inflammatory properties, and regulatory functions in glucose or lipid metabolism, several non-antidiabetic drugs have shown beneficial effects in improving liver histology, aminotransferase level, and metabolic parameters and reducing risks of hepatocellular carcinoma and mortality, without significant safety concerns, in patients with CLD. Although the effect as the centerpiece management in patients with CLD is not robust, the use of these non-antidiabetic drugs might be potentially beneficial as an adjuvant or combined treatment strategy.
Collapse
Affiliation(s)
- Han Ah Lee
- Departments of Internal Medicine, Ewha Womans University College of Medicine, Seoul, Korea
| | - Young Chang
- Department of Internal Medicine, Institute for Digestive Research, Digestive Disease Center, Soonchunhyang University College of Medicine, Seoul, Korea
| | - Pil Soo Sung
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- The Catholic University Liver Research Center, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Eileen L. Yoon
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea
| | - Hye Won Lee
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Yonsei Liver Center, Severance Hospital, Seoul, Korea
| | - Jeong-Ju Yoo
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Young-Sun Lee
- Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Jihyun An
- Department of Gastroenterology and Hepatology, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Korea
| | - Do Seon Song
- Department of Internal Medicine, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Young Youn Cho
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
| | - Seung Up Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Yonsei Liver Center, Severance Hospital, Seoul, Korea
| | - Yoon Jun Kim
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
19
|
Bian C, Zhang H, Gao J, Wang Y, Li J, Guo D, Wang W, Song Y, Weng Y, Ren H. SIRT6 regulates SREBP1c-induced glucolipid metabolism in liver and pancreas via the AMPKα-mTORC1 pathway. J Transl Med 2022; 102:474-484. [PMID: 34923569 DOI: 10.1038/s41374-021-00715-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/11/2021] [Accepted: 11/12/2021] [Indexed: 11/09/2022] Open
Abstract
The aim of this study was to determine the mechanism by which SIRT6 regulates glucolipid metabolism disorders. We detected histological and molecular changes in Sprague-Dawley rats as well as in BRL 3A and INS-1 cell lines subjected to overnutrition and starvation. SIRT6, SREBP1c, and glucolipid metabolism biomarkers were identified by fluorescence co-localization, real-time PCR, and western blotting. Gene silencing studies were performed. Recombinant SIRT6, AMPK agonist (AICAR), mTOR inhibitor (rapamycin), and liver X receptor (LXR) agonist (T0901317) were used to pre-treated in BRL 3A and INS-1 cells. Real-time PCR and western blotting were used to detect related proteins, and cell counting was utilized to detect proliferation. We obtained conflicting results; SIRT6 and SREBP1c appeared in both the liver and pancreas of high-fat and hungry rats. Recombinant SIRT6 alleviated the decrease in AMPKα and increase in mTORC1 (complex of mTOR, Raptor, and Rheb) caused by overnutrition. SIRT6 siRNA reversed the glucolipid metabolic disorders caused by the AMPK agonist and mTOR inhibitor but not by the LXR agonist. Taken together, our results demonstrate that SIRT6 regulates glycolipid metabolism through AMPKα-mTORC1 regulating SREBP1c in the liver and pancreas induced by overnutrition and starvation, independent of LXR.
Collapse
Affiliation(s)
- Che Bian
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Haibo Zhang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, Liaoning, China
| | - Jing Gao
- Department of Gerontology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yuxia Wang
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Jia Li
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Dan Guo
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Wei Wang
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Yuling Song
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Yang Weng
- Department of Gastroenterology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Huiwen Ren
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, Liaoning, China.
| |
Collapse
|
20
|
Aierken A, Li B, Liu P, Cheng X, Kou Z, Tan N, Zhang M, Yu S, Shen Q, Du X, Enkhbaatar BB, Zhang J, Zhang R, Wu X, Wang R, He X, Li N, Peng S, Jia W, Wang C, Hua J. Melatonin treatment improves human umbilical cord mesenchymal stem cell therapy in a mouse model of type II diabetes mellitus via the PI3K/AKT signaling pathway. Stem Cell Res Ther 2022; 13:164. [PMID: 35414044 PMCID: PMC9006413 DOI: 10.1186/s13287-022-02832-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 03/01/2022] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are promising candidates for tissue regeneration and disease treatment. However, long-term in vitro passaging leads to stemness loss of MSCs, resulting in failure of MSC therapy. This study investigated whether the combination of melatonin and human umbilical cord mesenchymal stem cells (hUC-MSCs) was superior to hUC-MSCs alone in ameliorating high-fat diet and streptozocin (STZ)-induced type II diabetes mellitus (T2DM) in a mouse model. METHODS Mice were divided into four groups: normal control (NC) group; T2DM group; hUC-MSCs treatment alone (UCMSC) group and pretreatment of hUC-MSCs with melatonin (UCMSC/Mel) group. RESULTS RNA sequence analysis showed that certain pathways, including the signaling pathway involved in the regulation of cell proliferation signaling pathway, were regulated by melatonin. The blood glucose levels of the mice in the UCMSC and UCMSC/Mel treatment groups were significantly reduced compared with the T2DM group without treatment (P < 0.05). Furthermore, hUC-MSCs enhance the key factor in the activation of the PI3K/Akt pathway in T2DM mouse hepatocytes. CONCLUSION The pretreatment of hUC-MSCs with melatonin partly boosted cell efficiency and thereby alleviated impaired glycemic control and insulin resistance. This study provides a practical strategy to improve the application of hUC-MSCs in diabetes mellitus and cytotherapy. Overview of the PI3K/AKT signaling pathway. (A) Underlying mechanism of UCMSC/Mel inhibition of hyperglycemia and insulin resistance T2DM mice via regulation of PI3K/AKT pathway. hUC-MSCs stimulates glucose uptake and improves insulin action thus should inhibition the clinical signs of T2DM, through activation of the p-PI3K/Akt signaling pathway and then regulates glucose transport through activating AS160. UCMSC/Mel increases p53-dependent expression of BCL2, and inhibit BAX and Capase3 protein activation. Leading to the decrease in apoptosis. (B) Melatonin modulated PI3K/AKT signaling pathway. Melatonin activated PI3K/AKT response pathway through binding to MT1and MT2 receptor. Leading to the increase in hUC-MSCs proliferation, migration and differentiation. → (Direct stimulatory modification); ┴ ( Direct Inhibitory modification); → ┤ (Multistep inhibitory modification); ↑ (Up regulate); ↓ (Down regulate); PI3K (Phosphoinositide 3-Kinase); AKT ( protein kinase B); PDK1 (Phosphoinositide-dependent protein kinase 1); IR, insulin receptor; GLUT4 ( glucose transporter type 4); ROS (reactive oxygen species); BCL-2 (B-cell lymphoma-2); PDK1 (phosphoinositide-dependent kinase 1) BAX (B-cell lymphoma-2-associated X protein); PCNA (Proliferating cell nuclear antigen); Cell cycle-associated proteins (KI67, cyclin A, cyclin E).
Collapse
Affiliation(s)
- Aili Aierken
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, YanglingShaanxi, 712100, China
| | - Balun Li
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, YanglingShaanxi, 712100, China
| | - Peng Liu
- Department of Endocrinology and Metabolism, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Xuedi Cheng
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, YanglingShaanxi, 712100, China
| | - Zheng Kou
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, YanglingShaanxi, 712100, China
| | - Ning Tan
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, YanglingShaanxi, 712100, China
| | - Mengfei Zhang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, YanglingShaanxi, 712100, China
| | - Shuai Yu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, YanglingShaanxi, 712100, China
| | - Qiaoyan Shen
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, YanglingShaanxi, 712100, China
| | - Xiaomin Du
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, YanglingShaanxi, 712100, China
| | - Bold Bayar Enkhbaatar
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, YanglingShaanxi, 712100, China
| | - Juqing Zhang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, YanglingShaanxi, 712100, China
| | - Rui Zhang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, YanglingShaanxi, 712100, China
| | - Xiaolong Wu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, YanglingShaanxi, 712100, China
| | - Ruibin Wang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, YanglingShaanxi, 712100, China
| | - Xin He
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, YanglingShaanxi, 712100, China
| | - Na Li
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, YanglingShaanxi, 712100, China
| | - Sha Peng
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, YanglingShaanxi, 712100, China
| | - Wenwen Jia
- Institute for Regenerative Medicine, National Stem Cell Translational Resource Center, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Congrong Wang
- Department of Endocrinology and Metabolism, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China.
| | - Jinlian Hua
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, YanglingShaanxi, 712100, China.
| |
Collapse
|
21
|
Malekpour-Dehkordi Z, Nourbakhsh M, Shahidi M, Sarraf N, Sharifi R. "Silymarin diminishes oleic acid-induced lipid accumulation in HepG2 cells by modulating the expression of endoplasmic reticulum stress markers". J Herb Med 2022. [DOI: 10.1016/j.hermed.2022.100565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
22
|
Salvoza N, Giraudi PJ, Tiribelli C, Rosso N. Natural Compounds for Counteracting Nonalcoholic Fatty Liver Disease (NAFLD): Advantages and Limitations of the Suggested Candidates. Int J Mol Sci 2022; 23:2764. [PMID: 35269912 PMCID: PMC8911502 DOI: 10.3390/ijms23052764] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 02/23/2022] [Accepted: 02/27/2022] [Indexed: 12/20/2022] Open
Abstract
The booming prevalence of nonalcoholic fatty liver disease (NAFLD) in adults and children will threaten the health system in the upcoming years. The "multiple hit" hypothesis is the currently accepted explanation of the complex etiology and pathophysiology of the disease. Some of the critical pathological events associated with the development of NAFLD are insulin resistance, steatosis, oxidative stress, inflammation, and fibrosis. Hence, attenuating these events may help prevent or delay the progression of NAFLD. Despite an increasing understanding of the mechanisms involved in NAFLD, no approved standard pharmacological treatment is available. The only currently recommended alternative relies on lifestyle modifications, including diet and physical activity. However, the lack of compliance is still hampering this approach. Thus, there is an evident need to characterize new therapeutic alternatives. Studies of food bioactive compounds became an attractive approach to overcome the reticence toward lifestyle changes. The present study aimed to review some of the reported compounds with beneficial properties in NAFLD; namely, coffee (and its components), tormentic acid, verbascoside, and silymarin. We provide details about their protective effects, their mechanism of action in ameliorating the critical pathological events involved in NAFLD, and their clinical applications.
Collapse
Affiliation(s)
- Noel Salvoza
- Fondazione Italiana Fegato—ONLUS, Area Science Park Basovizza, SS14 km 163.5, 34149 Trieste, Italy; (N.S.); (P.J.G.)
- Philippine Council for Health Research and Development, DOST Compound, Bicutan, Taguig 1631, Philippines
| | - Pablo J. Giraudi
- Fondazione Italiana Fegato—ONLUS, Area Science Park Basovizza, SS14 km 163.5, 34149 Trieste, Italy; (N.S.); (P.J.G.)
| | - Claudio Tiribelli
- Fondazione Italiana Fegato—ONLUS, Area Science Park Basovizza, SS14 km 163.5, 34149 Trieste, Italy; (N.S.); (P.J.G.)
| | - Natalia Rosso
- Fondazione Italiana Fegato—ONLUS, Area Science Park Basovizza, SS14 km 163.5, 34149 Trieste, Italy; (N.S.); (P.J.G.)
| |
Collapse
|
23
|
Chen J, Wan L, Zheng Q, Lan M, Zhang X, Li Y, Li B, Li L. Structural characterization and in vitro hypoglycaemic activity of glucomannan from Anemarrhena asphodeloides Bunge. Food Funct 2022; 13:1797-1807. [PMID: 35083996 DOI: 10.1039/d1fo03010h] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
A new polysaccharide (AABP-2B) was obtained from Anemarrhena asphodeloides Bunge after purification by gradient alcohol precipitation and DEAE-52 cellulose column chromatography. AABP-2B was confirmed to be a homogeneous polysaccharide with a molecular weight of 5800 Da and was composed of mannose and glucose at a molar ratio of 7.2 : 2.8. Structural analysis demonstrated that the backbone of AABP-2B was mainly composed of 4)-β-D-Manp-(1, 4,6)-β-D-Glcp-(1 and 3,6)-β-D-Manp-(1. The hypoglycaemic effect of AABP-2B was evaluated by its inhibition of α-glucosidase activities and insulin resistance in a HepG2 cell model. The results showed that AABP-2B displayed α-glucosidase inhibitory activities and could significantly improve glucose consumption by activating the IRS-1/PI3K/Akt signalling pathway in insulin-resistant HepG2 cells. Hence, AABP-2B may have potential as a functional food or medicine for diabetes therapy.
Collapse
Affiliation(s)
- Juncheng Chen
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Plant Protein Deep Processing, Ministry of Education, South China University of Technology, Guangzhou, 510640, China. .,International School of Public Health and One Health, Hainan Medical University, Haikou, Hainan 571199, China.
| | - Liting Wan
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Plant Protein Deep Processing, Ministry of Education, South China University of Technology, Guangzhou, 510640, China.
| | - Qingsong Zheng
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Plant Protein Deep Processing, Ministry of Education, South China University of Technology, Guangzhou, 510640, China.
| | - Meijuan Lan
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Plant Protein Deep Processing, Ministry of Education, South China University of Technology, Guangzhou, 510640, China.
| | - Xia Zhang
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Plant Protein Deep Processing, Ministry of Education, South China University of Technology, Guangzhou, 510640, China.
| | - Yuting Li
- School of Chemical Engineering and Energy Technology, Dongguan University of Technology, Dongguan, 523808, China
| | - Bing Li
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Plant Protein Deep Processing, Ministry of Education, South China University of Technology, Guangzhou, 510640, China.
| | - Lin Li
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Plant Protein Deep Processing, Ministry of Education, South China University of Technology, Guangzhou, 510640, China. .,School of Chemical Engineering and Energy Technology, Dongguan University of Technology, Dongguan, 523808, China
| |
Collapse
|
24
|
MacDonald-Ramos K, Michán L, Martínez-Ibarra A, Cerbón M. Silymarin is an ally against insulin resistance: A review. Ann Hepatol 2022; 23:100255. [PMID: 32950646 DOI: 10.1016/j.aohep.2020.08.072] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 08/10/2020] [Accepted: 08/12/2020] [Indexed: 02/07/2023]
Abstract
Silymarin is obtained from the Milk thistle plant Silybum marianum and has been used over the centuries to treat principally liver disease, although it has also been studied for its beneficial effects in cardioprotection, neuroprotection, immune modulation, and cancer among others. Importantly, silymarin's active component silybin is a flavonolignan that exhibits different activities such as; scavenger, anti-oxidant, anti-inflammatory, and recently revealed, insulin-sensitizing properties which have been explored in clinical trials in patients with insulin resistance. In this review, we summarize the most relevant research of silymarin's effect on lipid and carbohydrate metabolism, focusing the attention on insulin resistance, which is well known to play a crucial role in metabolic disease progression.
Collapse
Affiliation(s)
- Karla MacDonald-Ramos
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología "Isidro Espinosa de los Reyes" - Facultad de Química, Universidad Nacional Autónoma de México, CDMX, 11000, Mexico
| | - Layla Michán
- Facultad de Ciencias, Universidad Nacional Autónoma de México, CDMX, 04510, Mexico
| | - Alejandra Martínez-Ibarra
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología "Isidro Espinosa de los Reyes" - Facultad de Química, Universidad Nacional Autónoma de México, CDMX, 11000, Mexico
| | - Marco Cerbón
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología "Isidro Espinosa de los Reyes" - Facultad de Química, Universidad Nacional Autónoma de México, CDMX, 11000, Mexico.
| |
Collapse
|
25
|
Sun R, Xiao R, Lv P, Guo F, Gong Y, Yan M. Pink Lotus Essential Oil and Alleviates on Free Fatty Acid Induced Steatosis in HepG2 Cells via PI3K/Akt and NF-κB Pathways. J Oleo Sci 2022; 71:95-104. [PMID: 35013040 DOI: 10.5650/jos.ess21228] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Pink lotus essential oil (PLEO) is the volatile components extracted from lotus flowers and there are few relevant research. The purpose of this study was to observe the effect of PLEO on NAFLD in vitro model and its possible mechanism. The ingredients of PLEO were determined by gas chromatography-mass spectrometry (GS-MS) and its lipid-lowering and hepatoprotective activities were investigated. HepG2 cells were treated with free fatty acid (FFA) to establish a cell model of NAFLD. Cell viability was evaluated by 3-(4,5-dimethyl thiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) method. Total cholesterol (TC), triglyceride (TG), tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6) were determined by Enzyme-Linked Immune Sorbent Assay (ELISA). Oil red O staining was performed to observe the lipid accumulation in the HepG2 cells. Lipid metabolism enzymes including fatty acid synthase (FAS), acetyl-coA carboxylase (ACC), stearoyl-CoA desaturase 1 (SCD-1), and carnitine palmitoyltransferase-1 (CPT-1), insulin signaling pathways including phosphatidylinositol 3 kinase (PI3K) and protein kinase B Akt, inflammatory signaling pathways such as nuclear factor kappa-B (NF-κB), were determined by Western blotting. There were 46 components determined in PLEO with many terpenoids compounds. PLEO decreased TC and TG contents in the FFA-treated HepG2 cells. Furthermore, PLEO inhibited TNF-α, IL-6 and IL-1β excretion, decreased NF-κB, FAS, ACC and SCD-1 while increased phosphorylation of NF-κB, PI3K, Akt, and CPT-1 expression. It is the first time to reveal that PLEO alleviates FFA-induced steatosis in HepG2 cells by regulating lipid metabolism, inhibiting inflammatory response, and improving insulin sensitivity.
Collapse
Affiliation(s)
- Runzhou Sun
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology
| | - Ruixin Xiao
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology
| | - Pengfei Lv
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology
| | - Feifei Guo
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University
| | - Yanling Gong
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology
| | | |
Collapse
|
26
|
A comprehensive review on phytochemicals for fatty liver: are they potential adjuvants? J Mol Med (Berl) 2022; 100:411-425. [PMID: 34993581 DOI: 10.1007/s00109-021-02170-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 11/17/2021] [Accepted: 11/22/2021] [Indexed: 12/18/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is considered the hepatic manifestation of metabolic syndrome and, as such, is associated with obesity. With the current and growing epidemic of obesity, NAFLD is already considered the most common liver disease in the world. Currently, there is no official treatment for the disease besides weight loss. Although there are a few synthetic drugs currently being studied, there is also an abundance of herbal products that could also be used for treatment. With the World Health Organization (WHO) traditional medicine strategy (2014-2023) in mind, this review aims to analyze the mechanisms of action of some of these herbal products, as well as evaluate toxicity and herb-drug interactions available in literature.
Collapse
|
27
|
Hüttl M, Markova I, Miklankova D, Zapletalova I, Poruba M, Racova Z, Vecera R, Malinska H. The Beneficial Additive Effect of Silymarin in Metformin Therapy of Liver Steatosis in a Pre-Diabetic Model. Pharmaceutics 2021; 14:pharmaceutics14010045. [PMID: 35056941 PMCID: PMC8780287 DOI: 10.3390/pharmaceutics14010045] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/17/2021] [Accepted: 12/21/2021] [Indexed: 12/30/2022] Open
Abstract
The combination of plant-derived compounds with anti-diabetic agents to manage hepatic steatosis closely associated with diabetes mellitus may be a new therapeutic approach. Silymarin, a complex of bioactive substances extracted from Silybum marianum, evinces an antioxidative, anti-inflammatory, and hepatoprotective activity. In this study, we investigated whether metformin (300 mg/kg/day for four weeks) supplemented with micronized silymarin (600 mg/kg/day) would be effective in mitigating fatty liver disturbances in a pre-diabetic model with dyslipidemia. Compared with metformin monotherapy, the metformin-silymarin combination reduced the content of neutral lipids (TAGs) and lipotoxic intermediates (DAGs). Hepatic gene expression of enzymes and transcription factors involved in lipogenesis (Scd-1, Srebp1, Pparγ, and Nr1h) and fatty acid oxidation (Pparα) were positively affected, with hepatic lipid accumulation reducing as a result. Combination therapy also positively influenced arachidonic acid metabolism, including its metabolites (14,15-EET and 20-HETE), mitigating inflammation and oxidative stress. Changes in the gene expression of cytochrome P450 enzymes, particularly Cyp4A, can improve hepatic lipid metabolism and moderate inflammation. All these effects play a significant role in ameliorating insulin resistance, a principal background of liver steatosis closely linked to T2DM. The additive effect of silymarin in metformin therapy can mitigate fatty liver development in the pre-diabetic state and before the onset of diabetes.
Collapse
Affiliation(s)
- Martina Hüttl
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, 14021 Prague, Czech Republic; (I.M.); (D.M.); (H.M.)
- Correspondence: ; Tel.: +420-261-365-369
| | - Irena Markova
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, 14021 Prague, Czech Republic; (I.M.); (D.M.); (H.M.)
| | - Denisa Miklankova
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, 14021 Prague, Czech Republic; (I.M.); (D.M.); (H.M.)
| | - Iveta Zapletalova
- Department of Pharmacology, Faculty of Medicine and Dentistry, Palacky University, 77900 Olomouc, Czech Republic; (I.Z.); (M.P.); (Z.R.); (R.V.)
| | - Martin Poruba
- Department of Pharmacology, Faculty of Medicine and Dentistry, Palacky University, 77900 Olomouc, Czech Republic; (I.Z.); (M.P.); (Z.R.); (R.V.)
| | - Zuzana Racova
- Department of Pharmacology, Faculty of Medicine and Dentistry, Palacky University, 77900 Olomouc, Czech Republic; (I.Z.); (M.P.); (Z.R.); (R.V.)
| | - Rostislav Vecera
- Department of Pharmacology, Faculty of Medicine and Dentistry, Palacky University, 77900 Olomouc, Czech Republic; (I.Z.); (M.P.); (Z.R.); (R.V.)
| | - Hana Malinska
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, 14021 Prague, Czech Republic; (I.M.); (D.M.); (H.M.)
| |
Collapse
|
28
|
The Roles of Liver Inflammation and the Insulin Signaling Pathway in PM2.5 Instillation-Induced Insulin Resistance in Wistar Rats. DISEASE MARKERS 2021; 2021:2821673. [PMID: 34745386 PMCID: PMC8570885 DOI: 10.1155/2021/2821673] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 09/29/2021] [Indexed: 11/17/2022]
Abstract
To elucidate the mechanism of how the liver participates in PM2.5-caused insulin resistance. A novel Wistar rat model was developed in this study by instilling a suspension of lyophilized PM2.5 sample (2.5 mg/kg, 5 mg/kg, or 10 mg/kg) collected from the atmosphere. Systemic insulin resistance indicators, including serum fasting blood glucose (FBG), fasting insulin (FINS), Homeostatic Model Assessment for Insulin Resistance (HOMA-IR), and hemoglobin A1 (HbA1), were upregulated by the PM2.5 instillation. The area under the curve (AUCglu) calculated by intraperitoneal glucose tolerance testing (IPGTT) was also significantly greater in the PM2.5 instillation groups. Additionally, PM2.5 instillation was found to cause liver damage and inflammation. The serum levels of aspartate aminotransferase (AST), alanine aminotransferase (ALT), total bilirubin (TBIL), tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) were significantly elevated by PM2.5 instillation. PM2.5 also triggered IL-6 and TNF-α transcription but inhibited mRNA synthesis and suppressed signaling activation of the insulin-phosphoinositide 3-kinase- (PI3K-) Akt-glucose transporter 2 (GLUT2) pathway in the rat liver by reducing the ratio of phosphorylated Akt to phosphorylated insulin receptor substrate 1 (IRS-1). Thus, PM2.5-induced inflammation activation and insulin signaling inhibition in the rat liver contribute to the development of systemic insulin resistance.
Collapse
|
29
|
Zhang L, Xie Z, Yu H, Du H, Wang X, Cai J, Qiu Y, Chen R, Jiang X, Liu Z, Li Y, Chen T. TLR2 inhibition ameliorates the amplification effect of LPS on lipid accumulation and lipotoxicity in hepatic cells. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1429. [PMID: 34733981 PMCID: PMC8506759 DOI: 10.21037/atm-21-4012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/10/2021] [Indexed: 12/04/2022]
Abstract
Background Gut microbiome dysbiosis is related to the pathogenesis of nonalcoholic fatty liver disease (NAFLD), and the role of toll-like receptor 2 (TLR2) in its molecular mechanism is controversial. Here, we investigated the effects and mechanisms of Escherichia coli-derived lipopolysaccharide (LPS) on lipid accumulation and lipotoxicity in palmitic acid (PA)-treated L02 cell as an NAFLD cell model, and the role of TLR2 in this process. Methods Oil red O staining assay and free fatty acid (FFA) content test were performed to determine the effects of LPS on lipid accumulation in a PA-induced NAFLD cell model with or without TLR2 inhibition. The levels of IL-6 and TNF-α were measured to investigate inflammation conditions. Hoechst 33342 staining assay and Caspase-3 activity assay were used to test cell apoptosis, and the expression levels of proteins in the IRS1/PI3K/AKT signaling pathway, TLR2/MyD88/IKKα/NF-κB signaling pathway, and mitochondrion-dependent apoptotic signaling pathway were detected using Western blot. Results Lipid accumulation, pro-inflammatory cytokine release, and cell apoptosis with high levels were observed in the PA-induced NAFLD cell model, and LPS aggravated these processes. Whereas TLR2 inhibition could significantly ameliorate PA-induced and LPS-amplified lipid accumulation, inflammatory, and cell apoptosis, it had no significant effect on L02 cells treated with LPS alone. Conclusions These results were confirmed by activation or inhibition of the IRS1/PI3K/AKT signaling pathway, TLR2/MyD88/IKKα/NF-κB signaling pathway, and mitochondrion-dependent apoptotic signaling pathway, and were reflected by changes on their proteins expression. TLR2 is involved in PA-induced lipid accumulation and lipotoxicity in L02 cells, which could be aggravated by LPS, although LPS-induced amplification might not be through direct interaction with TLR2.
Collapse
Affiliation(s)
- Liting Zhang
- State Key Laboratory of Cryospheric Science, Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, China.,University of Chinese Academy of Sciences, Beijing, China.,Department of Infectious Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Zehui Xie
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Hongmiao Yu
- Children's Hospital and Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Haoxuan Du
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Xuqiao Wang
- School/Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Jiazheng Cai
- School/Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Yingfei Qiu
- School/Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Rui Chen
- School/Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Xiaofeng Jiang
- School/Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Zelin Liu
- School/Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Yi Li
- State Key Laboratory of Cryospheric Science, Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, China.,School/Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Tuo Chen
- State Key Laboratory of Cryospheric Science, Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
30
|
You Y, Chen L, Wu Y, Wang M, Lu H, Zhou X, Liu H, Fu Z, He Q, Ou J, Fu X, Liu Y, Kwan H, Liang D, Zhao X, Dai J. Silibinin Promotes Cell Proliferation Through Facilitating G1/S Transitions by Activating Drp1-Mediated Mitochondrial Fission in Cells. Cell Transplant 2021; 29:963689720950213. [PMID: 32830544 PMCID: PMC7563805 DOI: 10.1177/0963689720950213] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Heart, liver, and kidney, which are known as the essential organs for metabolism,
possess the unique ability to regulate the proliferation function of the body
against injury. Silibinin (SB), a natural polyphenolic flavonoid extracted from
traditional herb Silybum marianum L., has been used to protect
hepatocytes. Whether SB can regulate mitochondrial fission in normal cells and
the underlying mechanisms remain unclear. Here, we showed that SB markedly
promoted cell proliferation by facilitating G1/S transition via activating
dynamin-related protein 1 (Drp1), which in turn mediated mitochondrial fission
in these normal cells. SB dose-dependently increased the mitochondrial mass,
mtDNA copy number, cellular adenosine triphosphate production, mitochondrial
membrane potential, and reactive oxygen species in normal cells. Furthermore, SB
dose-dependently increased the expression of Drp1. Blocking Drp1 abolished
SB-induced mitochondrial fission. In conclusion, we demonstrate that SB promotes
cell proliferation through facilitating G1/S transition by activating
Drp1-mediated mitochondrial fission. This study suggests that SB is a
potentially useful herbal derivative for the daily prevention of various
diseases caused by impaired mitochondrial fission.
Collapse
Affiliation(s)
- Yanting You
- Department of Traditional Chinese Medicine, 70570Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Syndrome Laboratory of Integrated Chinese and Western Medicine, School of Chinese Medicine, 70570Southern Medical University, Guangzhou, Guangdong, China
| | - Liqian Chen
- Syndrome Laboratory of Integrated Chinese and Western Medicine, School of Chinese Medicine, 70570Southern Medical University, Guangzhou, Guangdong, China
| | - Yifen Wu
- Department of Oncology, Affiliated Dongguan People's Hospital, 70570Southern Medical University, Guangzhou, Guangdong, China
| | - Ming Wang
- Department of Traditional Chinese Medicine, 70570Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Hanqi Lu
- Syndrome Laboratory of Integrated Chinese and Western Medicine, School of Chinese Medicine, 70570Southern Medical University, Guangzhou, Guangdong, China
| | - Xinghong Zhou
- Syndrome Laboratory of Integrated Chinese and Western Medicine, School of Chinese Medicine, 70570Southern Medical University, Guangzhou, Guangdong, China
| | - Huaxi Liu
- Syndrome Laboratory of Integrated Chinese and Western Medicine, School of Chinese Medicine, 70570Southern Medical University, Guangzhou, Guangdong, China
| | - Zixuan Fu
- Syndrome Laboratory of Integrated Chinese and Western Medicine, School of Chinese Medicine, 70570Southern Medical University, Guangzhou, Guangdong, China
| | - Qiuxing He
- Syndrome Laboratory of Integrated Chinese and Western Medicine, School of Chinese Medicine, 70570Southern Medical University, Guangzhou, Guangdong, China
| | - Jinying Ou
- Traditional Chinese Pharmacological Laboratory, School of Traditional Chinese Medicine, 70570Southern Medical University, Guangzhou, Guangdong, China
| | - Xiuqiong Fu
- School of Chinese Medicine, 26679Hong Kong Baptist University, Hong Kong, China
| | - Yanyan Liu
- Department of Traditional Chinese Medicine, 70570Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Hiuyee Kwan
- School of Chinese Medicine, 26679Hong Kong Baptist University, Hong Kong, China
| | - Donghui Liang
- Department of Traditional Chinese Medicine, 70570Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaoshan Zhao
- Syndrome Laboratory of Integrated Chinese and Western Medicine, School of Chinese Medicine, 70570Southern Medical University, Guangzhou, Guangdong, China
| | - Jiaojiao Dai
- Syndrome Laboratory of Integrated Chinese and Western Medicine, School of Chinese Medicine, 70570Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
31
|
Yang L, Liu Q, Zhang H, Wang Y, Li Y, Chen S, Song G, Ren L. Silibinin improves nonalcoholic fatty liver by regulating the expression of miR‑122: An in vitro and in vivo study. Mol Med Rep 2021; 23:335. [PMID: 33760189 PMCID: PMC7974327 DOI: 10.3892/mmr.2021.11974] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 01/12/2021] [Indexed: 02/06/2023] Open
Abstract
Silibinin is a flavonoid that improves fatty liver and insulin resistance. To elucidate the effect of silibinin on lipid deposition and the potential molecular mechanism, the present study conducted in vivo and in vitro experiments. In the in vivo experiments, mice were randomly divided into control, high‑fat and silibinin groups, while HepG2 cells were randomly divided into control, palmitic acid intervention and silibinin intervention groups. The mRNA, protein and miR‑122 expression associated with hepatic lipid metabolism were detected in each group. The results demonstrated that silibinin reduced the triglyceride content, miR‑122 expression and the mRNA and protein expressions of fatty acid synthase (FAS) and acetyl‑CoA carboxylase (ACC). Silibinin increased the mRNA and protein expression of carnitine palmitoyl transferase 1A (CPT1A). In the present study, HepG2 cells cultured with palmitate were treated with silibinin following overexpression of micro RNA (miR) 122. The results demonstrated that the mRNA and protein expression of FAS and ACC was increased, while that of CPT1A was decreased. Therefore, it could be deduced that silibinin improved lipid metabolism by reducing the expression of miR‑122 and inhibiting the expression of miR‑122 may be a new therapeutic target to improve fatty liver disease.
Collapse
Affiliation(s)
- Liying Yang
- Endocrinology Department, Hebei General Hospital, Shijiazhuang, Hebei 050000, P.R. China
| | - Qianqian Liu
- Endocrinology Department, Hebei General Hospital, Shijiazhuang, Hebei 050000, P.R. China
| | - He Zhang
- Endocrinology Department, Hebei General Hospital, Shijiazhuang, Hebei 050000, P.R. China
| | - Yichao Wang
- Endocrinology Department, Hebei General Hospital, Shijiazhuang, Hebei 050000, P.R. China
| | - Yang Li
- Endocrinology Department, Hebei General Hospital, Shijiazhuang, Hebei 050000, P.R. China
| | - Shuchun Chen
- Endocrinology Department, Hebei General Hospital, Shijiazhuang, Hebei 050000, P.R. China
| | - Guangyao Song
- Endocrinology Department, Hebei General Hospital, Shijiazhuang, Hebei 050000, P.R. China
| | - Luping Ren
- Endocrinology Department, Hebei General Hospital, Shijiazhuang, Hebei 050000, P.R. China
| |
Collapse
|
32
|
New insight and potential therapy for NAFLD: CYP2E1 and flavonoids. Biomed Pharmacother 2021; 137:111326. [PMID: 33556870 DOI: 10.1016/j.biopha.2021.111326] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 01/21/2021] [Accepted: 01/25/2021] [Indexed: 12/13/2022] Open
Abstract
Over the years, the prevalence of nonalcoholic fatty liver disease (NAFLD) has increased year by year; however, due to its complicated pathogenesis, there is no effective treatment so far. It is reported that Cytochrome P450 2E1 (CYP2E1) plays an indispensable role in the development of NAFLD, and numerous studies have shown that flavonoids have a hepatoprotective effect and can exert a beneficial effect on NAFLD by regulating the activity of CYP2E1. Therefore, flavonoids may become effective drugs for the treatment of NAFLD in the future. This prompted us to review the research progress of the pathological mechanism of NAFLD and the impact of CYP2E1 activity changes during the pathological process, and to summarize the protective effect of flavonoids against CYP2E1 activity.
Collapse
|
33
|
Tighe SP, Akhtar D, Iqbal U, Ahmed A. Chronic Liver Disease and Silymarin: A Biochemical and Clinical Review. J Clin Transl Hepatol 2020; 8:454-458. [PMID: 33447529 PMCID: PMC7782115 DOI: 10.14218/jcth.2020.00012] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/20/2020] [Accepted: 09/08/2020] [Indexed: 02/07/2023] Open
Abstract
Chronic liver disease (CLD) is an under-recognized epidemic that continues to increase in prevalence and is a major health concern. Silymarin, the active compound of Silybum marianum (Milk thistle), has historically been used in CLD. A significant barrier to silymarin use is its poor bioavailability. Attempts at improving the bioavailability of silymarin have led to a better understanding of formulation methods, pharmacokinetics, dosing, and associated drug interactions. Clinically, silymarin exerts its hepatoprotective effects through antioxidative, antifibrotic, anti-inflammatory, antitoxin, and anticancerous mechanisms of actions. Despite the use of silymarin being extensively studied in alcoholic liver disease, metabolic-associated fatty liver disease, viral hepatitis, and drug-induced liver injury, the overall efficacy of silymarin remains unclear and more research is warranted to better elucidate the role of silymarin in CLD, specifically regarding its anti-inflammatory effects. Here, we review the current biochemical and clinical evidence regarding silymarin in CLD.
Collapse
Affiliation(s)
- Sean P. Tighe
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Daud Akhtar
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Umair Iqbal
- Department of Gastroenterology and Hepatology, Geisinger Commonwealth School of Medicine, Danville, PA, USA
- *Correspondence to: Umair Iqbal, Department of Gastroenterology and Hepatology, Geisinger Commonwealth School of Medicine, Danville, PA 17821, USA. Tel: +1-570-271-6211, E-mail:
| | - Aijaz Ahmed
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
34
|
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
|
35
|
Li X, Wang Y, Xing Y, Xing R, Liu Y, Xu Y. Changes of gut microbiota during silybin-mediated treatment of high-fat diet-induced non-alcoholic fatty liver disease in mice. Hepatol Res 2020; 50:5-14. [PMID: 31661720 DOI: 10.1111/hepr.13444] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/17/2019] [Accepted: 10/04/2019] [Indexed: 12/12/2022]
Abstract
AIM Gut microbiota are involved in the pathogenesis of non-alcoholic fatty liver disease (NAFLD). Silybin (Sil), a naturally occurring hepatoprotective agent, is widely used for treating NAFLD. Whether Sil affects gut microbiota during its actions in treating NAFLD is unknown. We aimed to examine the effect of Sil on intestinal flora dysbiosis induced by a high-fat diet (HFD). METHODS After 10 weeks of feeding normal chow diet or HFD, mice were given a daily gavage for 8 weeks. Cecal contents were harvested for study of short-chain fatty acids, bile acids, and gut microbiota alteration. RESULTS Sil showed protective effects against dietary-induced obesity and liver steatosis; accordingly, gut microbiota composition changed. At the phylum level, compared with the HFD group, mice in the Sil-treated group had significantly lower levels of Firmicutes, and the ratio of Firmicutes-to-Bacteroidetes was lower (P < 0.05). At the genus level, the Sil-treated group have significantly lower levels of Lachnoclostridium, Lachnospiraceae_UCG-006, and Mollicutes_RF9, which were reported to be potentially related to diet-induced obesity, and increased levels of Blautia (P < 0.05), Akkermansia (P < 0.05), and Bacteroides (P < 0.05), which are known to have a beneficial effect on improving NAFLD. Sil also showed an inhibitory effect on well-known beneficial bacteria, such as Alloprevotella and Lactobacillus. Furthermore, the production of acetate, propionate, and butyrate increased, whereas the generation of formate and conversion of cytotoxic secondary metabolites (lithocholic acid and deoxy-cholic acid) decreased in mice treated with Sil. CONCLUSIONS Sil might have beneficial effects on ameliorating NAFLD and mediating HFD-induced change of gut microbiota composition, followed by major changes in secondary metabolites, such as short-chain fatty acids and bile acids.
Collapse
Affiliation(s)
- Xiuxia Li
- Eastern Hospital, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Yanping Wang
- Eastern Hospital, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Yilan Xing
- Eastern Hospital, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Renxin Xing
- Eastern Hospital, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Yongsheng Liu
- Eastern Hospital, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Yinsheng Xu
- Eastern Hospital, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| |
Collapse
|
36
|
Zheng X, Zhao MG, Jiang CH, Sheng XP, Yang HM, Liu Y, Yao XM, Zhang J, Yin ZQ. Triterpenic acids-enriched fraction from Cyclocarya paliurus attenuates insulin resistance and hepatic steatosis via PI3K/Akt/GSK3β pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 66:153130. [PMID: 31790897 DOI: 10.1016/j.phymed.2019.153130] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 10/16/2019] [Accepted: 10/29/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is the most prevalent form of chronic liver diseases. Cyclocarya paliurus (C. paliurus), an edible and medicinal plant in Chinese folk, has been demonstrated to ameliorate diabetes, obesity and lipid metabolism disorders. However, its effects on NAFLD and its potential molecular mechanism have not been clearly expounded. PURPOSE The present study was designed to explore the therapeutic potential of triterpenic acids-enriched fraction from C. paliurus (CPT), as well as its underlying mechanism in vivo and in vitro models of NAFLD. METHODS The metabolic effects and possible molecular mechanism of CPT were examined using HepG2 cells and primary hepatocytes (isolated from C57BL/6 J mice) models of fatty liver induced by palmitic acid (PA) and a high fat diet mouse model. RESULTS In high fat diet-induced C57BL/6 J mice, CPT significantly reduced liver weight index, serum alanine transaminase (ALT), aspartate transaminase (AST), triacylglycerol (TG), total cholesterol (TC) and hepatic TG, TC levels. Moreover, CPT dramatically decreased the contents of blood glucose, insulin, and insulin resistance (HOMA-IR) index. Meanwhile, CPT significantly increased the tyrosine phosphorylation level of IRS and the uptake of 2-deoxyglucose (2DG) in PA-induced HepG2 cells and primary hepatocytes fatty liver models. Furthermore, in PA-induced HepG2 cells and primary hepatocytes, CPT significantly decreased the number of lipid droplets and intracellular TG content. In addition, mechanism investigation showed that CPT increased the phosphorylation of phosphoinositide 3-kinase (PI3K), protein kinase B (Akt) and glycogen synthase-3β (GSK3β) in vivo and in vitro models, which were abrogated by PI3K inhibitor LY294002 in vitro models. CONCLUSION These findings indicate that CPT may exert the therapeutic effects on NAFLD via regulating PI3K/Akt/GSK3β pathway.
Collapse
Affiliation(s)
- Xian Zheng
- Department of TCMs Pharmaceuticals & State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China; Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210009, China; Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210009, China
| | - Meng-Ge Zhao
- Department of TCMs Pharmaceuticals & State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China; Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210009, China; Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210009, China
| | - Cui-Hua Jiang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210009, China; Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210009, China
| | - Xue-Ping Sheng
- Department of TCMs Pharmaceuticals & State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China; Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210009, China; Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210009, China
| | - Hui-Min Yang
- Department of TCMs Pharmaceuticals & State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China; Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210009, China; Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210009, China
| | - Yao Liu
- Department of TCMs Pharmaceuticals & State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China; Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210009, China; Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210009, China
| | - Xiao-Ming Yao
- Clinical Laboratory, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu 210028, China
| | - Jian Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210009, China; Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210009, China.
| | - Zhi-Qi Yin
- Department of TCMs Pharmaceuticals & State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
37
|
Liu W, Sun H, Zhou Y, Li Y, Qin Y, Li R, Chen Y, Yu L, Zhao M, Zhang W, Xu Y. Goat Milk Consumption Ameliorates Abnormalities in Glucose Metabolism and Enhances Hepatic and Skeletal Muscle AMP-Activated Protein Kinase Activation in Rats Fed with High-Fat Diets. Mol Nutr Food Res 2019; 63:e1900703. [PMID: 31750605 DOI: 10.1002/mnfr.201900703] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 11/13/2019] [Indexed: 12/15/2022]
Abstract
SCOPE Diabetes endangers health and causes serious economic impediment. The aim of this study is to identify the effects of goat milk consumption on glucose metabolism of rats with high-fat (HF) diet. METHODS AND RESULTS Sixty male Sprague Dawley rats are divided into five groups and fed with different diets for 24 weeks: goat-milk-based HF diet (GHF group; goat milk powder+HF diet), cow-milk-based HF diet (CHF group; cow milk powder+HF diet), HF diet, HF diet plus acarbose (HF+A group; acarbose+HF diet), and chow diet (CD group). Fasting glucose in GHF-fed rats are lower than HF-fed rats on weeks 16 and 20. GHF-fed rats display improved insulin sensitivity in oral glucose and insulin tolerance tests. Compared with HF-fed rats, glycated hemoglobin and triglycerides in GHF-fed rats are lower and high-density lipoprotein level is higher. AMP-activated protein kinase activation (AMPK) in the liver and skeletal muscle is higher in GHF rats than HF rats. Phosphoenolpyruvate carboxykinase and glucose 6-phosphatase protein levels in the liver are lower and hexokinase 2 protein level in the skeletal muscle is higher in GHF rats compared with HF rats. CONCLUSION Goat milk consumption can ameliorate abnormalities in glucose metabolism, and AMPK pathway in the liver and skeletal muscle plays an important role in the process.
Collapse
Affiliation(s)
- Wei Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Xueyuan Road 38, Haidian, Beijing, 100083, China
| | - Han Sun
- Research and Development Department, Heilongjiang Feihe Dairy Co., Ltd., C-16, 10A Jiuxianqiao Rd., Chaoyang, Beijing, 100015, China
| | - Yalin Zhou
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Xueyuan Road 38, Haidian, Beijing, 100083, China
| | - Yong Li
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Xueyuan Road 38, Haidian, Beijing, 100083, China
| | - Yong Qin
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Xueyuan Road 38, Haidian, Beijing, 100083, China
| | - Ruijun Li
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Xueyuan Road 38, Haidian, Beijing, 100083, China
| | - Yuhan Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Xueyuan Road 38, Haidian, Beijing, 100083, China
| | - Lanlan Yu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Xueyuan Road 38, Haidian, Beijing, 100083, China
| | - Mengya Zhao
- Research and Development Department, Heilongjiang Feihe Dairy Co., Ltd., C-16, 10A Jiuxianqiao Rd., Chaoyang, Beijing, 100015, China
| | - Wei Zhang
- Research and Development Department, Heilongjiang Feihe Dairy Co., Ltd., C-16, 10A Jiuxianqiao Rd., Chaoyang, Beijing, 100015, China
| | - Yajun Xu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Xueyuan Road 38, Haidian, Beijing, 100083, China.,Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Xueyuan Road 38, Haidian, Beijing, 100083, China
| |
Collapse
|
38
|
Pharmacological Therapy of Non-Alcoholic Fatty Liver Disease: What Drugs Are Available Now and Future Perspectives. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16224334. [PMID: 31703268 PMCID: PMC6888162 DOI: 10.3390/ijerph16224334] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/01/2019] [Accepted: 11/03/2019] [Indexed: 12/25/2022]
Abstract
The non-alcoholic fatty liver disease (NAFLD) is rapidly becoming the most common cause of chronic liver disease as well as the first cause of liver transplantation. NAFLD is commonly associated with metabolic syndrome (MetS), and this is the most important reason why it is extremely difficult to treat this disease bearing in mind the enormous amount of interrelationships between the liver and other systems in maintaining the metabolic health. The treatment of NAFLD is a key point to prevent NASH progression to advanced fibrosis, to prevent cirrhosis and to prevent the development of its hepatic complications (such as liver decompensation and HCC) and even extrahepatic one. A part of the well-known healthy effect of diet and physical exercise in this setting it is important to design the correct pharmaceutical strategy in order to antagonize the progression of the disease. In this regard, the current review has the scope to give a panoramic view on the possible pharmacological treatment strategy in NAFLD patients.
Collapse
|
39
|
Li Y, Wang C, Lu J, Huang K, Han Y, Chen J, Yang Y, Liu B. PPAR δ inhibition protects against palmitic acid-LPS induced lipidosis and injury in cultured hepatocyte L02 cell. Int J Med Sci 2019; 16:1593-1603. [PMID: 31839747 PMCID: PMC6909814 DOI: 10.7150/ijms.37677] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 08/22/2019] [Indexed: 01/18/2023] Open
Abstract
Background: Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease, and its pathogenesis and mechanism are intricate. In the present study, we aimed to evaluate the role of PPAR δ in LPS associated NAFLD and to investigate the signal transduction pathways underlying PPAR δ treatment in vitro. Material and Methods: L02 cells were exposed to palmitic acid (PA) and/or LPS in the absence or presence of PPAR δ inhibition and/or activation. Results: LPS treatment markedly increased lipid deposition, FFA contents, IL-6 and TNF-α levels, and cell apoptosis in PA treatment (NAFLD model). PPAR δ inhibition protects L02 cells against LPS-induced lipidosis and injury. Conversely, the result of PPAR δ activation showed the reverse trend. LPS+PA treatment group significantly decreases the relative expression level of IRS-1, PI3K, AKT, phosphorylation of AKT, TLR-4, MyD88, phosphorylation of IKKα, NF-κB, Bcl-2 and increases the relative expression level of Bax, cleaved caspase 3 and cleaved caspase 8, compared with the cells treated with NAFLD model. PPAR δ inhibition upregulated the related proteins' expression level in insulin resistance and inflammation pathway and downregulated apoptotic relevant proteins. Instead, PPAR δ agonist showed the reverse trend. Conclusion: Our data show that PPAR δ inhibition reduces steatosis, inflammation and apoptosis in LPS-related NAFLD damage, in vitro. PPAR δ may be a potential therapeutic implication for NAFLD.
Collapse
Affiliation(s)
- Yi Li
- School/Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Chenwei Wang
- School/Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Jiyuan Lu
- School/Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Ke Huang
- School/Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Yu Han
- College of Life Science & Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Junlin Chen
- School/Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Yan Yang
- Department of Endocrinology, Gansu Provincial Hospital, Lanzhou, China
| | - Bin Liu
- School/Hospital of Stomatology, Lanzhou University, Lanzhou, China
| |
Collapse
|
40
|
Yan H, Li T, Wang Y, Li H, Xu J, Lu X. Insulin-like growth factor binding protein 7 accelerates hepatic steatosis and insulin resistance in non-alcoholic fatty liver disease. Clin Exp Pharmacol Physiol 2019; 46:1101-1110. [PMID: 31397492 DOI: 10.1111/1440-1681.13159] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 08/04/2019] [Accepted: 08/06/2019] [Indexed: 12/19/2022]
Abstract
An association between increased insulin-like growth factor binding protein-7 (IGFBP7) expression and insulin resistance in metabolic diseases has been reported. However, the role and molecular mechanism of IGFBP-7 in non-alcoholic fatty liver disease (NAFLD) remains largely unknown. Therefore, the potential function of IGFBP7 in the pathological progression of NAFLD was explored in this investigation. For in vivo experiments, an animal model of NAFLD was established in C57BL/6 mice by feeding a high-fat diet (HFD), and IGFBP7 was knocked down by injecting adeno-associated adenovirus (AAV)-mediated short-hairpin (sh)-IGFBP7 into the liver. We found that AAV-sh-IGFBP7 treatment significantly alleviated hepatocyte injury and inhibited hepatic lipid accumulation by reducing lipogenesis-associated gene expression. Furthermore, downregulation of IGFBP7 markedly ameliorated IR and restored impaired insulin signalling by elevating the phosphorylation levels of IRS-1, Akt and GSK3β in HFD-treated mice. Similar results were also confirmed by an in vitro study in a palmitic acid (PA)-stimulated HepG2 cell model. In conclusion, our study demonstrates that IGFBP7 contributes to hepatic steatosis and insulin resistance in NAFLD development, which might serve as a novel therapeutic agent for the treatment of NAFLD.
Collapse
Affiliation(s)
- Hua Yan
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Department of Gerontology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Ting Li
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yatao Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hong Li
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jingyuan Xu
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaolan Lu
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
41
|
Zhuang Y, Xing C, Cao H, Zhang C, Luo J, Guo X, Hu G. Insulin resistance and metabonomics analysis of fatty liver haemorrhagic syndrome in laying hens induced by a high-energy low-protein diet. Sci Rep 2019; 9:10141. [PMID: 31300671 PMCID: PMC6626135 DOI: 10.1038/s41598-019-46183-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 06/24/2019] [Indexed: 12/31/2022] Open
Abstract
Fatty liver haemorrhagic syndrome (FLHS) is a widespread metabolic disease in laying hens that causes a decrease in egg production and even death. Insulin resistance is a major contributor to the pathogenesis of nonalcoholic fatty liver disease. However, the relationship between FLHS and the insulin resistance mechanisms underlying FLHS is not well elucidated. Therefore, we established an FLHS model induced by feeding a high-energy low-protein diet. In the current study, we found that the fasting glucose and insulin concentrations were elevated in the FLHS group compared with the control group during the experimental period. The results of the oral glucose tolerance test (OGTT) and insulin sensitivity test (IST) showed a high level of insulin resistance in the FLHS model. InsR, 4EBP-1, Glut-1 and Glut-3 mRNA expression were decreased, and TOR, S6K1, and FOXO1 were elevated (P < 0.05). Metabolomic analysis with GC/MS identified 46 differentially expressed metabolites between these two groups, and of these, 14 kinds of metabolism molecules and 32 kinds of small metabolism molecules were decreased (P < 0.05). Further investigation showed that glucose, lipid and amino acid metabolism blocks in the progression of FLHS by GO functional and pathway analysis. Overall, these results suggest that insulin resistance participated in FLHS; comprehensively, metabolites participated in the dysregulated biological process.
Collapse
Affiliation(s)
- Yu Zhuang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang, 330045, Jiangxi, P.R. China
| | - Chenghong Xing
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang, 330045, Jiangxi, P.R. China
| | - Huabin Cao
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang, 330045, Jiangxi, P.R. China
| | - Caiying Zhang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang, 330045, Jiangxi, P.R. China
| | - Junrong Luo
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang, 330045, Jiangxi, P.R. China
| | - Xiaoquan Guo
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang, 330045, Jiangxi, P.R. China
| | - Guoliang Hu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang, 330045, Jiangxi, P.R. China.
| |
Collapse
|
42
|
Liu Y, Xu W, Zhai T, You J, Chen Y. Silibinin ameliorates hepatic lipid accumulation and oxidative stress in mice with non-alcoholic steatohepatitis by regulating CFLAR-JNK pathway. Acta Pharm Sin B 2019; 9:745-757. [PMID: 31384535 PMCID: PMC6664044 DOI: 10.1016/j.apsb.2019.02.006] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 12/11/2018] [Accepted: 01/11/2019] [Indexed: 02/07/2023] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is a chronic metabolic syndrome and the CFLAR-JNK pathway can reverse the process of NASH. Although silibinin is used for the treatment of NASH in clinical, its effect on CFLAR-JNK pathway in NASH remains unclear. This study aimed to investigate the effect of silibinin on CFLAR-JNK pathway in NASH models both in vivo and in vitro. The in vivo study was performed using male C57BL/6 mice fed with methionine- choline-deficient diet and simultaneously treated with silibinin for 6 weeks. The in vitro study was performed by using mouse NCTC-1469 cells which were respectively pretreated with oleic acid plus palmitic acid, and adenovirus-down Cflar for 24 h, then treated with silibinin for 24 h. After the drug treatment, the key indicators involved in CFLAR-JNK pathway including hepatic injury, lipid metabolism and oxidative stress were determined. Silibinin significantly activated CFLAR and inhibited the phosphorylation of JNK, up-regulated the mRNA expression of Pparα, Fabp5, Cpt1α, Acox, Scd-1, Gpat and Mttp, reduced the activities of serum ALT and AST and the contents of hepatic TG, TC and MDA, increased the expression of NRF2 and the activities of CAT, GSH-Px and HO-1, and decreased the activities and expression of CYP2E1 and CYP4A in vivo. These effects were confirmed by the in vitro experiments. Silibinin prevented NASH by regulating CFLAR-JNK pathway, and thereby on one hand promoting the β-oxidation and efflux of fatty acids in liver to relieve lipid accumulation, and on the other hand inducing antioxidase activity (CAT, GSH-Px and HO-1) and inhibiting pro-oxidase activity (CYP2E1 and CYP4A) to relieve oxidative stress.
Collapse
Key Words
- 2-NBDG, 2-(N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl) amino)-2-deoxyglucose
- ALT, alanine aminotransferase
- AST, aspartate aminotransferase
- Acox, acyl-coenzyme A oxidase X
- Akt, serine–threonine protein kinase
- CAT, catalase
- CFLAR
- CFLAR, caspase 8 and Fas-associated protein with death domain-like apoptosis regulator
- CYP2E1, cytochrome P450 2E1
- CYP4A, cytochrome P450 4A
- Cpt1α, carnitine palmitoyl transferase 1α
- Fabp5, fatty acid-binding proteins 5
- GSH-Px, glutathione peroxidase
- Gpat, glycerol-3-phosphate acyltransferase
- HE, hematoxylin–eosin
- HO-1, heme oxygenase 1
- IR, insulin resistance
- IRS1, insulin receptor substrate 1
- JNK, c-Jun N-terminal kinase
- Lipid accumulation
- MAPK, mitogen-activated protein kinase
- MCD, methionine- and choline-deficient
- MCS, methionine- and choline-sufficient
- MDA, malondialdehyde
- MT, Masson–Trichrome
- Mttp, microsomal triglyceride transfer protein
- NAFLD, non-alcoholic fatty liver disease
- NASH
- NASH, nonalcoholic steatohepatitis
- NF-κB, nuclear factor κB
- NRF2, nuclear factor erythroid 2-related factor 2
- OA, oleic acid
- ORO, oil red O
- Oxidation stress
- PA, palmitic acid
- PI3K, phosphatidylinositol 3-hydroxy kinase
- Pnpla3, phospholipase domain containing 3
- Pparα, peroxisome proliferator activated receptor α
- SD, Sprague–Dawley
- Scd-1, stearoyl-coenzyme A desaturase-1
- Silibinin
- Srebp-1c, sterol regulatory element binding protein-1C
- TC, total cholesterol
- TG, triglyceride
- pIRS1, phosphorylation of insulin receptor substrate 1
- pJNK, phosphorylation of c-Jun N-terminal kinase
Collapse
Affiliation(s)
| | | | | | | | - Yong Chen
- Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, Hubei University, Wuhan 430062, China
| |
Collapse
|
43
|
Li B, Fang J, He T, Yin S, Yang M, Cui H, Ma X, Deng J, Ren Z, Hu Y, Ye G, Zhang M, Geng Y, Gou L, Zuo Z. Resistin up-regulates LPL expression through the PPARγ-dependent PI3K/AKT signaling pathway impacting lipid accumulation in RAW264.7 macrophages. Cytokine 2019; 119:168-174. [PMID: 30925325 DOI: 10.1016/j.cyto.2019.03.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 03/17/2019] [Accepted: 03/21/2019] [Indexed: 12/19/2022]
Abstract
Resistin is a cysteine-rich cytokine, which has been indicated as a mediator of insulin resistance and inflammation. Previous studies demonstrated that lipoprotein lipase (LPL) was an important enzyme that could mediate lipid accumulation in macrophages. Additionally, the intracellular molecules phosphatidylinositol 3-kinase (PI3K)/serine-threonine protein kinase (AKT)/peroxisome proliferator-activated receptor (PPARγ) were supposed to be involved in the lipid accumulation process in cells. However, it remains unclear whether resistin was correlated with the dysregulation of lipid metabolism in macrophages. The present study investigated that resistin could up-regulate the expression of LPL and increase the contents of intracellular triglyceride (TG) and total cholesterol (TC) in RAW264.7 macrophages. In addition, intracellular molecules PI3K, AKT and PPARγ were significantly up-regulated and activated in resitin-stimulated RAW264.7 macrophages (P < 0.05). In contrast, the effects of resistin on RAW264.7 macrophages could be abrogated by specific inhibitors for LPL (LPL-siRNA) and PI3K/AKT signaling pathway (LY294002). All together, this study demonstrated that resistin could up-regulate the expression of LPL and induce lipid accumulation in RAW264.7 macrophages. More importantly, the PPARγ-dependent PI3K/AKT signaling pathway was relevant to the lipid accumulation process in resistin-stimulated macrophages.
Collapse
Affiliation(s)
- Bi Li
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Huimin road 211, 611130, China; Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Chengdu Research Base of Giant Panda Breeding, Chengdu, Sichuan 610081, China
| | - Jing Fang
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Huimin road 211, 611130, China
| | - Tingting He
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Huimin road 211, 611130, China
| | - Sirui Yin
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Huimin road 211, 611130, China
| | - Mingxian Yang
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Huimin road 211, 611130, China; Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Chengdu Research Base of Giant Panda Breeding, Chengdu, Sichuan 610081, China
| | - Hengmin Cui
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Huimin road 211, 611130, China
| | - Xiaoping Ma
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Huimin road 211, 611130, China
| | - Junliang Deng
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Huimin road 211, 611130, China
| | - Zhihua Ren
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Huimin road 211, 611130, China
| | - Yanchun Hu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Huimin road 211, 611130, China
| | - Gang Ye
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Huimin road 211, 611130, China
| | - Ming Zhang
- College of Animal Science & Technology, Sichuan Agricultural University, Huimin road 211, 611130, China
| | - Yi Geng
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Huimin road 211, 611130, China
| | - Liping Gou
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Huimin road 211, 611130, China
| | - Zhicai Zuo
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Huimin road 211, 611130, China.
| |
Collapse
|
44
|
Yang Y, Li J, Wei C, He Y, Cao Y, Zhang Y, Sun W, Qiao B, He J. Amelioration of nonalcoholic fatty liver disease by swertiamarin in fructose-fed mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 59:152782. [PMID: 31005808 DOI: 10.1016/j.phymed.2018.12.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 10/09/2018] [Accepted: 12/09/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is the most prevalent chronic liver disease. Swertia bimaculata (Sieb. et Zucc.) Hook. Thoms.ex Clarke, a glabrous or procumbent perennial herb, is a traditional herb medicine. Swertiamarin, a secoiridoid glycoside, is a representative ingredient in this medical plant crude extract and shows antidiabetic and antihyperlipidaemic activities and protective effect against hepatic injury. PURPOSE The present study aimed to determine whether swertiamarin can attenuate NAFLD in fructose-fed mice. METHODS Healthy male mice freely drank water containing 10% fructose for 12 consecutive weeks, whereas animals in those swertiamarin tested groups received different doses of swertiamarin (25, 50 and 100 mg/kg) by intragastric administration once a day from the ninth week to the twelfth week. RESULTS At the end of the experiment, fructose-fed mice administrated with swertiamarin showed low levels of serum glucose, triglycerides, uric acid, alanine aminotransferase and aspartate transaminase. Histological examinations suggested the alleviation of hepatic ballooning degeneration and steatosis by swertiamarin treatment. Moreover, swertiamarin administration mitigated hepatic oxidative stress along with decreases of hepatic pro-inflammation cytokines, which was associated with decrease of hepatic xanthine oxidase (XO) activity and enhancements of anti-oxidant defense system enzymes, as well as activation of nuclear factor E2-related factor 2 (Nrf2) in fructose-fed mice. In addition, swertiamarin down-regulated expression of sterol-regulatory element-binding protein-1 (SREBP-1), fatty acid synthase (FAS) and acetyl-CoA carboxylase 1 (ACC1) in liver of fructose-fed mice. CONCLUSION The present study demonstrates that swertiamarin alleviates NAFLD and metabolic alterations in fructose-fed mice.
Collapse
Affiliation(s)
- Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, Xi'an 710069, China; Biomedicine Key Laboratory of Shaanxi Province, College of Life Science, Northwest University, Xi'an 710069, China
| | - Jing Li
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, Xi'an 710069, China; Biomedicine Key Laboratory of Shaanxi Province, College of Life Science, Northwest University, Xi'an 710069, China.
| | - Cong Wei
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, Xi'an 710069, China; Biomedicine Key Laboratory of Shaanxi Province, College of Life Science, Northwest University, Xi'an 710069, China
| | - Ying He
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, Xi'an 710069, China; Biomedicine Key Laboratory of Shaanxi Province, College of Life Science, Northwest University, Xi'an 710069, China
| | - Yixin Cao
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, Xi'an 710069, China; Biomedicine Key Laboratory of Shaanxi Province, College of Life Science, Northwest University, Xi'an 710069, China
| | - Yongmin Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, Xi'an 710069, China; Biomedicine Key Laboratory of Shaanxi Province, College of Life Science, Northwest University, Xi'an 710069, China; Sorbonne Université, Institut Parisien de Chimie Moléculaire, CNRS UMR 8232, 4 place Jussieu, Paris 75005, France
| | - Wenji Sun
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, Xi'an 710069, China; Biomedicine Key Laboratory of Shaanxi Province, College of Life Science, Northwest University, Xi'an 710069, China
| | - Boling Qiao
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, Xi'an 710069, China; Biomedicine Key Laboratory of Shaanxi Province, College of Life Science, Northwest University, Xi'an 710069, China
| | - Jiao He
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, Xi'an 710069, China; Biomedicine Key Laboratory of Shaanxi Province, College of Life Science, Northwest University, Xi'an 710069, China
| |
Collapse
|
45
|
Xu Y, Chen D, Lin XX, Zhao Q, Guo J, Chen LJ, Zhang W, Xiao J, Lian GH, Peng SF, Guo D, Yang H, Obianom O, Shu Y, Chen Y. The LRP6 functional mutation rs2302685 contributes to individual susceptibility to alcoholic liver injury related to the Wnt/β-catenin-TCF1-CYP2E1 signaling pathway. Arch Toxicol 2019; 93:1679-1695. [PMID: 30976847 DOI: 10.1007/s00204-019-02447-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 04/09/2019] [Indexed: 10/27/2022]
Abstract
Low-density lipoprotein receptor-related protein 6 (LRP6) is an important coreceptor in the Wnt/β-catenin upstream signaling pathway. Rs2302685 is a common functional mutation of LRP6 that has been previously associated with reduced alcoholic liver injury among alcoholic liver disease (ALD) patients, and the present research was designed to study the underlying mechanisms of that finding. A total of 107 ALD patients and 138 non-ALD patients were recruited from hospitalized alcoholics in China. Their venous blood samples were collected for DNA extraction and genotyped using Sequenom MassARRAY. We found that the rs2302685 mutation, which impaired the function of LRP6, was present in higher frequency among alcoholics with ALD than those without ALD. We also conducted a mouse model experiment in which LRP6(+/-) knockdown mice and LRP6(+/+) wild-type mice received daily intragastric doses of ethanol (2.4 g/kg) as well as a larger dose of ethanol (4 g/kg) every 7 days for 28 days. The mouse blood and liver specimens were subsequently collected for laboratory analysis, and cell experiments were performed to compare the inhibition, activation, over-expression, and siRNA of LRP6 in the treatment versus the control HL7702 cells. Expression of the targeted molecules was detected by real-time PCR or western blot analysis. Stably transfected cells with pRL3-CYP2E1 vector were used to further study the underlying mechanisms. The total bile acid (TBA), direct bilirubin, total bilirubin (TBIL), aspartate aminotransferase (AST), mitochondrial aspartate aminotransferase, and AST/ALT values were significantly lower in carriers of the rs2302685 mutation than in the wild-type patients, by 63.4, 60.6, 82.1, 44.8, 45.7, and 21.4%, respectively. Compared to the LRP6(+/+) wild-type mice, the LRP6(+/-) knockdown mice had lower ALT, TBIL, TBA, and ALB/GLO values, as well reduced liver tissue damage, in accordance with their reduced expressions of LRP6, β-catenin, and CYP2E1. In HL7702 cells exposed to ethanol, AST, ALT, lipid accumulation, and ROS generation decreased in cells that were treated with LRP6 inhibitors or siRNA but increased in cells treated with LRP6 activators or over-expressed LRP6. TCF1 was the transcriptional factor most likely to connect the LRP6-Wnt/β-catenin signaling pathway to the regulation of CYP2E1. We concluded that the LRP6 functional mutation rs2302685 contributes to individual differences in susceptibility to alcoholic liver injury related to the Wnt/β-catenin-TCF1-CYP2E1 signaling pathway.
Collapse
Affiliation(s)
- Ying Xu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.,Institute of Clinical Pharmacology, Central South University, Changsha, 410078, Hunan, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
| | - Dan Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.,Institute of Clinical Pharmacology, Central South University, Changsha, 410078, Hunan, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
| | - Xiu-Xian Lin
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.,Institute of Clinical Pharmacology, Central South University, Changsha, 410078, Hunan, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
| | - Qing Zhao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.,Institute of Clinical Pharmacology, Central South University, Changsha, 410078, Hunan, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
| | - Jing Guo
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.,Institute of Clinical Pharmacology, Central South University, Changsha, 410078, Hunan, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
| | - Li-Jie Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.,Institute of Clinical Pharmacology, Central South University, Changsha, 410078, Hunan, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.,Institute of Clinical Pharmacology, Central South University, Changsha, 410078, Hunan, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
| | - Jian Xiao
- Department of Hepatology and Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.,Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Guang-Hui Lian
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Shi-Fang Peng
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Dong Guo
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD, 21201, USA
| | - Hong Yang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD, 21201, USA
| | - Obinna Obianom
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD, 21201, USA
| | - Yan Shu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD, 21201, USA
| | - Yao Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China. .,Institute of Clinical Pharmacology, Central South University, Changsha, 410078, Hunan, China. .,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, People's Republic of China. .,National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
| |
Collapse
|
46
|
Xu F, Yang J, Negishi H, Sun Y, Li D, Zhang X, Hayashi T, Gao M, Ikeda K, Ikejima T. Silibinin decreases hepatic glucose production through the activation of gut-brain-liver axis in diabetic rats. Food Funct 2019; 9:4926-4935. [PMID: 30178798 DOI: 10.1039/c8fo00565f] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Silibinin, a flavonolignan derived from milk thistle (Silybum marianum), has been revealed to have a beneficial effect on improving diabetes-impaired glycemic control. However, the underlying mechanism is still unclear. In the present study, to evaluate whether the gut-brain-liver axis, an important neural pathway for the control of hepatic glucose production, is involved in silibinin-regulated glucose homeostasis, the expression of glucagon-like peptide-1 receptor (GLP1R) in the duodenum, activation of neurons in the nucleus of the solitary tract (NTS), as well as glycogen accumulation and expression of gluconeogenic enzymes in the livers of diabetic SHRSP·Z-Leprfa/IzmDmcr (SP·ZF) rats with 4-week oral administration of silibinin (100 and 300 mg kg-1 day-1) were evaluated. Common hepatic branch vagotomy was further conducted in high-fat diet/streptozotocin (HFD/STZ)-induced diabetic SD rats to confirm the role of the gut-brain-liver axis in silibinin-improved glycemic control. The results revealed a significant inhibition of fasting blood glucose after SP·ZF rats were administrated with silibinin for 4 weeks. The expression of GLP1R in the duodenum and the activation of neurons in the NTS increased, while hepatic glucose production decreased on silibinin administration. However, the hypoglycemic effect of silibinin was reversed by common hepatic branch vagotomy in diabetic SD rats. Our study suggested that silibinin may be useful as a potential functional food ingredient against diabetes by triggering the gut-brain-liver axis.
Collapse
Affiliation(s)
- Fanxing Xu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, P.R. China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Sciacqua A, Perticone M, Tripepi G, Addesi D, Cassano V, Maio R, Sesti G, Perticone F. Metabolic and vascular effects of silybin in hypertensive patients with high 1-h post-load plasma glucose. Intern Emerg Med 2019; 14:77-84. [PMID: 30232738 DOI: 10.1007/s11739-018-1951-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 09/13/2018] [Indexed: 11/28/2022]
Abstract
Hypertensive patients with normal glucose tolerance (NGT) but 1-h post-load plasma glucose ≥ 155 mg/dl (1-h high), during an oral glucose tolerance test (OGTT), show higher insulin resistance and multiple target organ damages. Experimental and clinical studies demonstrate that silybin presents anti-inflammatory and metabolic effects, improving insulin resistance and endothelial dysfunction. This study aims to evaluate the effects of the complex silybin-vitamin E and phospholipids on inflammatory, metabolic and vascular parameters in NGT 1-h high hypertensive patients. This is a pilot, single arm, interventional, longitudinal study enrolling 50 Caucasian NGT 1-h high hypertensive outpatients, 27 men and 23 women, age range 42-60 years (mean + SD = 52 ± 7). After 6 months of silybin intake, there is a significant improvement in metabolic profile. The glucose response during OGTT significantly improves (AUCglucose0-120 309.6 ± 63.4 at baseline vs 254.6 ± 35.5 at the follow-up, ∆ = - 55, 95% CI from - 67 to - 43, p < 0.0001), so as insulin response (AUCinsulin0-120 238.2 ± 99.1 vs 159.3 ± 44.9, ∆ = - 78.9, 95% CI from - 100.0 to - 57.8, p < 0.0001), in accordance with the increase of insulin sensitivity index Matsuda. Silybin intake is associated with a significant reduction of both clinical and central systolic blood pressure, with betterment in clinical and central pulse pressure and reduction of arterial stiffness parameters. In conclusion, this study demonstrates that silybin may improve the metabolic aspect and vascular damage in NGT 1-h high hypertensive patients who are at higher metabolic and cardiovascular risk. Thus, in these patients, silybin might strengthen the effect of antihypertensive drugs giving further cardiovascular protection.
Collapse
Affiliation(s)
- Angela Sciacqua
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Campus Universitario di Germaneto, V.le Europa, 88100, Catanzaro, Italy.
| | - Maria Perticone
- Department of Experimental and Clinical Medicine, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Giovanni Tripepi
- CNR-IFC, Istituto di Fisiologia Clinica, Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, Reggio Calabria, Italy
| | - Desire Addesi
- Internal Medicine Unit, Pugliese-Ciaccio Hospital, Catanzaro, Italy
| | - Velia Cassano
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Campus Universitario di Germaneto, V.le Europa, 88100, Catanzaro, Italy
| | - Raffaele Maio
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Campus Universitario di Germaneto, V.le Europa, 88100, Catanzaro, Italy
| | - Giorgio Sesti
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Campus Universitario di Germaneto, V.le Europa, 88100, Catanzaro, Italy
| | - Francesco Perticone
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Campus Universitario di Germaneto, V.le Europa, 88100, Catanzaro, Italy
| |
Collapse
|
48
|
Silibinin ameliorates diabetic nephropathy via improving diabetic condition in the mice. Eur J Pharmacol 2018; 845:24-31. [PMID: 30597130 DOI: 10.1016/j.ejphar.2018.12.031] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 12/11/2018] [Accepted: 12/20/2018] [Indexed: 02/02/2023]
Abstract
Diabetic nephropathy (DN) is a major cause of end-stage renal disease and one of the most severe diabetic complications. However, there is lack of effective treatments for DN and the underlying mechanisms of the renal injury remain unclear. In current study, we evaluated the effects of silibinin on DN and further explored the underlying mechanisms. We administrated silibinin to db/db mice for 10 weeks. Then we monitored the diabetic metabolic parameters, kidney function, oxidative stress and AKT signaling pathway in db/db mice. Administration of silibinin to db/db mice improved diabetic condition, as evidenced by the decrease of body weight, HbAc1level and serum insulin level in db/db mice. Silibinin prevented kidney injury and attenuated oxidative stress in db/db mice. Silibinin activated AKT signaling pathway and decreased the levels of p-GSK-3β, Bax and cleaved caspase-3. Silibinin ameliorates diabetic nephropathy by activating the AKT signaling pathway.
Collapse
|
49
|
Li H, Yu L, Zhao C. Dioscin attenuates high‑fat diet‑induced insulin resistance of adipose tissue through the IRS‑1/PI3K/Akt signaling pathway. Mol Med Rep 2018; 19:1230-1237. [PMID: 30483735 DOI: 10.3892/mmr.2018.9700] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 10/10/2018] [Indexed: 11/05/2022] Open
Abstract
Insulin resistance, as a common metabolic disorder, may be caused by diet‑induced obesity. The aim of the present study is to investigate the effects of dioscin on regulating insulin resistance of adipose tissue induced by a high‑fat diet (HFD). An animal model was established successfully using C57BL/6J mice with high‑fat feeding, followed by treatment with 5, 10 and 20 mg/kg dioscin through gavage for 18 weeks, and randomly divided into a control group, a HFD model group and a dioscin group treated with 5, 10 and 20 mg/kg/day dioscin for 12 weeks. Histopathological changes in adipose tissues were examined using hematoxylin and eosin staining. Biochemical parameters of the serum were also monitored, including glucose, insulin, total triglyceride, homeostasis model assessment of insulin resistance (HOMA‑IR) and adipose insulin resistance (Adipo‑IR) levels. Expression of the mRNA and associated proteins of the insulin receptor substrate 1 (IRS‑1)/phosphoinositide 3‑kinase (PI3K)/protein kinase B (Akt) pathways were determined using reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) and western blot analysis, respectively. HOMA‑IR and Adipo‑IR values of mice fed with a HFD were significantly higher compared with those in the control group (P<0.01). However, dioscin administration significantly decreased HOMA‑IR and Adipo‑IR values in a dose‑dependent manner (P<0.05), suggesting the effects of dioscin on attenuating insulin resistance. RT‑qPCR results indicated that the associated genes of the IRS‑1/PI3K/Akt pathway were significantly downregulated by HFD compared with the control group (P<0.05), while dioscin significantly increased the expression of those genes compared with the control group (P<0.05). Similarly, the significant increase in phosphorylated (p‑)IRS‑1/IRS‑1 (P<0.05) and p‑Akt/Akt (P<0.05) values were substantially reversed by dioscin treatment. Dioscin pronouncedly mitigated insulin resistance in adipose tissues through the IRS‑1/PI3K/Akt pathway and has potential to be used as a novel therapeutic agent for the therapy of HFD‑induced insulin resistance in adipose tissue.
Collapse
Affiliation(s)
- Haijuan Li
- Department of Clinical Nutrition, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Lianzhi Yu
- Health Check Centre, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Changsheng Zhao
- Department of Nutrition, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| |
Collapse
|
50
|
Teng W, Yin W, Zhao L, Ma C, Huang J, Ren F. Resveratrol metabolites ameliorate insulin resistance in HepG2 hepatocytes by modulating IRS-1/AMPK. RSC Adv 2018; 8:36034-36042. [PMID: 35558476 PMCID: PMC9088716 DOI: 10.1039/c8ra05092a] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 09/26/2018] [Indexed: 11/30/2022] Open
Abstract
Resveratrol (trans-3,5,4′-trihydroxystilbene, RSV), a naturally occurring biologically active polyphenol has been observed to induce numerous beneficial effects in diabetic animals and humans. However, its protective effects are somewhat controversial due to low bioavailability and rapid clearance rate. Therefore, we in this study have tried to investigate if its main metabolites, RSV-3-O-glucuronide (R3G) and RSV-4-O-glucuronide (R4G) could ameliorate insulin resistance, similar to RSV in insulin-resistant HepG2 cells. Herein, we first established an insulin-resistant cell model by treating HepG2 cells with 1 × 10−6 mol L−1 insulin for 24 h. Subsequently, the effects of R3G and R4G on insulin resistance inhibition were evaluated in HepG2 cells. Interestingly, our data indicated that R3G and R4G treatment improved cellular glucose uptake and glycogen synthesis contents, and blocked generation of intracellular reactive oxygen species (ROS). Additionally, R3G and R4G also modulated insulin signaling and improved insulin sensitivity by modulating the IRS-1/AMPK signaling pathway. Taken together, our data provided a significant new insight into the effects and molecular mechanism of R3G and R4G on ameliorating insulin resistance in HepG2 cells. Overall, our data supported the hypothesis that despite low bioavailability in vivo, RSV biological effects could be mediated through its metabolites. RSV metabolites R3G and R4G protected HepG2 cell from insulin resistance by improving glucose uptake and glycogen synthesis, along with inhibiting ROS generation and modulating the RS-1/AMPK signaling pathway.![]()
Collapse
Affiliation(s)
- Wendi Teng
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University P.O. Box 287, No. 17 Qinghua East Road Beijing 100083 China +86-10-62736344.,Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Municipality, College of Food Science & Nutritional Engineering, China Agricultural University Beijing China
| | - Wenjing Yin
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University P.O. Box 287, No. 17 Qinghua East Road Beijing 100083 China +86-10-62736344.,Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Municipality, College of Food Science & Nutritional Engineering, China Agricultural University Beijing China
| | - Liang Zhao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University P.O. Box 287, No. 17 Qinghua East Road Beijing 100083 China +86-10-62736344.,Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Municipality, College of Food Science & Nutritional Engineering, China Agricultural University Beijing China.,Beijing Higher Institution Engineering Research Center of Animal Product, College of Food Science & Nutritional Engineering, China Agricultural University Beijing China
| | - Changwei Ma
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University P.O. Box 287, No. 17 Qinghua East Road Beijing 100083 China +86-10-62736344.,Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Municipality, College of Food Science & Nutritional Engineering, China Agricultural University Beijing China.,Beijing Higher Institution Engineering Research Center of Animal Product, College of Food Science & Nutritional Engineering, China Agricultural University Beijing China
| | - Jiaqiang Huang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University P.O. Box 287, No. 17 Qinghua East Road Beijing 100083 China +86-10-62736344.,Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Municipality, College of Food Science & Nutritional Engineering, China Agricultural University Beijing China.,Beijing Higher Institution Engineering Research Center of Animal Product, College of Food Science & Nutritional Engineering, China Agricultural University Beijing China
| | - Fazheng Ren
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University P.O. Box 287, No. 17 Qinghua East Road Beijing 100083 China +86-10-62736344.,Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Municipality, College of Food Science & Nutritional Engineering, China Agricultural University Beijing China.,Beijing Higher Institution Engineering Research Center of Animal Product, College of Food Science & Nutritional Engineering, China Agricultural University Beijing China
| |
Collapse
|