1
|
Bhardwaj M, Mazumder PM. An insight on the additive impact of type 2 diabetes mellitus and nonalcoholic fatty liver disease on cardiovascular consequences. Mol Biol Rep 2025; 52:169. [PMID: 39873861 DOI: 10.1007/s11033-025-10249-0] [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: 04/29/2024] [Accepted: 01/10/2025] [Indexed: 01/30/2025]
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) and type 2 diabetes mellitus (T2DM) are associated with a multifactorial complicated aetiology that is often coexisting and has a strong and distinct connection with cardiovascular diseases (CVDs). In order to accomplish effective and appropriate therapeutic strategies, a deeper understanding of the bidirectional interaction between NAFLD patients, NAFLD patients with T2DM, and NAFLD patients with CVDs is required to control the concomitant rise in prevalence of these conditions worldwide. This article also aims to shed light on the epidemiology and mechanisms behind the relationship between T2DM, NAFLD and the related cardiovascular consequences. METHOD Literature was collected from PubMed, Medline, Embase, Web of Science and Google scholar from inception to June, 2024. For surveying literature different combinations and formats of terms including NAFLD, NASH, T2DM and CVDs were used. RESULTS In the recent decade, clinical and epidemiological studies have been conducted and provide strong evidence that NAFLD is closely linked with CVD progression along with associated morbidity and mortality in both patients with and without T2DM. Several mechanistic approaches contribute to cardiovascular consequences and abnormalities in cardiac biomarkers in T2DM and NAFLD patients, including adipose tissue malfunction, mitochondrial dysfunction, the microbiota, genetic and epigenetic alterations contributing to insulin resistance, glucotoxicity and lipotoxicity. CONCLUSION The study reveals a complex interplay between diabetes, hepatic and cardiovascular complications, leading to significant morbidity and mortality in diabetic and NAFLD patients. This pandemic necessitates further research to identify mitigating variables and develop effective treatment approaches.
Collapse
Affiliation(s)
- Monika Bhardwaj
- Department of Pharmaceutical Sciences & Technology, BIT Mesra, Ranchi, 835215, India
| | - Papiya Mitra Mazumder
- Department of Pharmaceutical Sciences & Technology, BIT Mesra, Ranchi, 835215, India.
| |
Collapse
|
2
|
Reda D, Elshopakey GE, Albukhari TA, Almehmadi SJ, Refaat B, Risha EF, Mahgoub HA, El-Boshy ME, Abdelhamid FM. Vitamin D3 alleviates nonalcoholic fatty liver disease in rats by inhibiting hepatic oxidative stress and inflammation via the SREBP-1-c/ PPARα-NF-κB/IR-S2 signaling pathway. Front Pharmacol 2023; 14:1164512. [PMID: 37261280 PMCID: PMC10228732 DOI: 10.3389/fphar.2023.1164512] [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: 02/12/2023] [Accepted: 05/02/2023] [Indexed: 06/02/2023] Open
Abstract
Introduction: Nonalcoholic fatty liver disease (NAFLD) is a chronic disease characterized by fat deposits in liver cells, which can lead to hepatitis and fibrosis. This study attempted to explore the protective effect of vitamin D3 (VitD) against NAFLD. Methods: Adult male albino rats were randomized into four separate groups: the negative control group was fed a standard rat chow; the positive group received a high-fat diet (20%) and 25% fructose water (NAFLD); the VitD control group was intramuscularly treated with VitD (1,000 IU/kg BW) 3 days per week for 10 weeks; and the NAFLD group was treated with VitD therapy. Biochemical and hepatic histological analyses were performed. Hepatic oxidative stress and inflammatory conditions were also studied. Hepatic expression of sterol regulatory element-binding protein 1-c (SREBP-1-c), peroxisome proliferator-activated receptor alpha (PPAR-α), and insulin receptor substrate-2 was analyzed by quantitative real-time polymerase chain reaction. Results and discussion: The NAFLD rats exhibited elevated terminal body weight, hepatic injury markers, dyslipidemia, glucose intolerance, and insulin resistance. Moreover, the NAFLD rats had increased SREBP-1-c expression and reduced PPAR-α and IRS-2 expressions. Histological analysis showed hepatic steatosis and inflammation in the NAFLD group. In contrast, VitD administration improved the serum biochemical parameters and hepatic redox status in NAFLD rats. Also, VitD treatment ameliorated hepatic inflammation and steatosis in the NAFLD group by decreasing the expression of SREBP-1-c and increasing the expression of PPAR-α. Overall, these results suggest that VitD could have a protective effect against NAFLD and its associated complication.
Collapse
Affiliation(s)
- Doha Reda
- Clinical Pathology Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Gehad E. Elshopakey
- Clinical Pathology Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Talat A. Albukhari
- Department of Haematology and Immunology, Faculty of Medicine, Umm Alqura University, Makkah, Saudi Arabia
| | - Samah J. Almehmadi
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Bassem Refaat
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Engy F. Risha
- Clinical Pathology Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Hebatallah A. Mahgoub
- Pathology Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Mohamed E. El-Boshy
- Clinical Pathology Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Fatma M. Abdelhamid
- Clinical Pathology Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
3
|
Yu L, Zhao R, Wang C, Zhang C, Chu C, Zhao J, Zhang H, Zhai Q, Chen W, Zhang H, Tian F. Effects of garlic supplementation on non-alcoholic fatty liver disease: A systematic review and meta-analysis of randomized controlled trials. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
4
|
Brown RCC, Jegatheesan DK, Conley MM, Mayr HL, Kelly JT, Webb L, Barnett A, Staudacher HM, Burton NW, Isbel NM, Macdonald GA, Campbell KL, Coombes JS, Keating SE, Hickman IJ. U-DECIDE: Utilising technology for Diet & Exercise Change In complex chronic conditions across Diverse Environments: Protocol for a Randomised Controlled Trial (Preprint). JMIR Res Protoc 2022; 11:e37556. [PMID: 35900834 PMCID: PMC9377441 DOI: 10.2196/37556] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/11/2022] [Accepted: 05/13/2022] [Indexed: 11/16/2022] Open
Abstract
Background The metabolic syndrome is common across many complex chronic disease groups. Advances in health technology have provided opportunities to support lifestyle interventions. Objective The purpose of this study is to test the feasibility of a health technology-assisted lifestyle intervention in a patient-led model of care. Methods The study is a single-center, 26-week, randomized controlled trial. The setting is specialist kidney and liver disease clinics at a large Australian tertiary hospital. The participants will be adults with a complex chronic condition who are referred for dietetic assessment and display at least one feature of the metabolic syndrome. All participants will receive an individualized assessment and advice on diet quality from a dietitian, a wearable activity monitor, and standard care. Participants randomized to the intervention group will receive access to a suite of health technologies from which to choose, including common base components (text messages) and optional components (online and mobile app–based nutrition information, an online home exercise program, and group-based videoconferencing). Exposure to the optional aspects of the intervention will be patient-led, with participants choosing their preferred level of engagement. The primary outcome will be the feasibility of delivering the program, determined by safety, recruitment rate, retention, exposure uptake, and telehealth adherence. Secondary outcomes will be clinical effectiveness, patient-led goal attainment, treatment fidelity, exposure demand, and participant perceptions. Primary outcome data will be assessed descriptively and secondary outcomes will be assessed using an analysis of covariance. This study will provide evidence on the feasibility of the intervention in a tertiary setting for patients with complex chronic disease exhibiting features of the metabolic syndrome. Results The study was funded in 2019. Enrollment has commenced and is expected to be completed by June 2022. Data collection and follow up are expected to be completed by December 2022. Results from the analyses based on primary outcomes are expected to be submitted for publication by June 2023. Conclusions The study will test the implementation of a health technology–assisted lifestyle intervention in a tertiary outpatient setting for a diverse group of patients with complex chronic conditions. It is novel in that it embeds patient choice into intervention exposure and will inform health service decision-makers in regards to the feasibility of scale and spread of technology-assisted access to care for a broader reach of specialist services. Trial Registration Australian New Zealand Clinical Trial Registry ACTRN12620001282976; https://www.anzctr.org.au/Trial/Registration/TrialReview.aspx?id=378337 International Registered Report Identifier (IRRID) DERR1-10.2196/37556
Collapse
Affiliation(s)
- Riley C C Brown
- School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, Australia
| | - Dev K Jegatheesan
- Department of Nephrology, Princess Alexandra Hospital, Brisbane, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Marguerite M Conley
- Department of Nutrition and Dietetics, Princess Alexandra Hospital, Brisbane, Australia
| | - Hannah L Mayr
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
- Department of Nutrition and Dietetics, Princess Alexandra Hospital, Brisbane, Australia
- Nutrition and Dietetics Research Group, Bond University, Robina, Australia
| | - Jaimon T Kelly
- Centre for Online Health, The University of Queensland, Brisbane, Australia
- Centre for Health Services Research, The University of Queensland, Brisbane, Australia
| | - Lindsey Webb
- Department of Nutrition and Dietetics, Princess Alexandra Hospital, Brisbane, Australia
| | - Amandine Barnett
- Centre for Online Health, The University of Queensland, Brisbane, Australia
- Centre for Health Services Research, The University of Queensland, Brisbane, Australia
| | - Heidi M Staudacher
- Food & Mood Centre, Institute for Mental and Physical Health and Clinical Translation, Deakin University, Melbourne, Australia
| | - Nicola W Burton
- School of Applied Psychology, Griffith University, Brisbane, Australia
| | - Nicole M Isbel
- Department of Nephrology, Princess Alexandra Hospital, Brisbane, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Graeme A Macdonald
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
- Department of Gastroenterology and Hepatology, Princess Alexandra Hospital, Brisbane, Australia
| | - Katrina L Campbell
- Healthcare Excellence and Innovation, Metro North Health, Brisbane, Australia
- Menzies Health Institute Queensland, Griffith University, Brisbane, Australia
| | - Jeff S Coombes
- School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, Australia
| | - Shelley E Keating
- School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, Australia
| | - Ingrid J Hickman
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
- Department of Nutrition and Dietetics, Princess Alexandra Hospital, Brisbane, Australia
| |
Collapse
|
5
|
Elabd EY, El-Beik DEDMS, Bayoumy EM, Saleh M, Ezzat WM, Siddik RI, Mohamed AE, Sayed MM, Ali MO. Diagnostic Value of Acyl-Ghrelin in Type 2 Diabetic Patients with Non-alcoholic Fatty Liver Disease. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2021.7548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND: Nonalcoholic fatty liver disease (NAFLD) has become the leading cause of chronic liver disease worldwide. Type 2 diabetes (T2D) is described as one of the most significant risk factor for developing NAFLD, non-alcoholic steatohepatitis, and advanced cirrhosis. Liver biopsy cannot be used routinely to diagnose NAFLD. Therefore, it is critically urgent to develop a simple non-invasive test.
AIM: This study examined fasting Acyl-Ghrelin (AG) as a non-invasive biomarker to accurately diagnose NAFLD in diabetic patients.
PATIENTS AND METHODS: Sixty-one patients with T2D were divided into a test group with NAFLD, and a control group without NAFLD. Secondary causes of fatty liver, chronic viral hepatitis, and drug-induced liver damage were excluded from the study. Anthropometric measurements, lipid profile, fasting blood sugar (FBS), liver enzyme activities, and fasting AG levels were collected. Data management and analysis were performed using statistical package for social sciences version 20.
RESULTS: Fasting AG level (pg/ml) in the test group (56.1 ± 10.7) was increased, but not statically significant compared with the control group (37.8 ± 9.3), p > 0.05. However, significant metabolic changes were observed in body weight, waist circumference, FBS, alanine transaminase, and aspartate transaminase between test and control groups. The mean values in the test group are 93.2 ± 14.5, 115.4 ± 7.6, 144.2 ± 25.9, 21.1 ± 5.7, and 32.3 ± 2.1. While the mean values are 87.7 ± 7.3, 95 ± 3.8, 123.7 ± 20.7, 18.6 ± 5, and 20 ± 7, respectively, in the control group.
CONCLUSIONS: While elevated AG levels alone were not significant, elevated AG levels plus other parameters of liver damage and obesity were associated with the diagnosis of NAFLD. However, more studies are needed to consider elevated AG as a diagnostic marker in NAFLD patients with T2D.
Collapse
|
6
|
Song C, Lv W, Li Y, Nie P, Lu J, Geng Y, Heng Z, Song L. Alleviating the effect of quinoa and the underlying mechanism on hepatic steatosis in high-fat diet-fed rats. Nutr Metab (Lond) 2021; 18:106. [PMID: 34922572 PMCID: PMC8684231 DOI: 10.1186/s12986-021-00631-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 11/26/2021] [Indexed: 02/08/2023] Open
Abstract
Background Nonalcoholic fatty liver disease (NAFLD) is considered the hepatic component of metabolic syndrome and has attracted widespread attention due to its increased prevalence. Daily dietary management is an effective strategy for the prevention of NAFLD. Quinoa, a nutritious pseudocereal, is abundant in antioxidative bioactive phytochemicals. In the present study, the effects of different amounts of quinoa on the progression of NAFLD and the related molecular mechanism were investigated. Methods Male SD rats were simultaneously administered a high fat diet (HF) and different amounts of quinoa (equivalent to 100 g/day and 300 g/day of human intake, respectively). After 12 weeks of the intervention, hepatic TG (triglyceride) and TC (total cholesterol) as well as serum antioxidative parameters were determined, and hematoxylin–eosin staining (H&E) staining was used to evaluate hepatic steatosis. Differential metabolites in serum and hepatic tissue were identified using UPLC-QTOF-MSE. The mRNA expression profile was investigated using RNA-Seq and further verified using real-time polymerase chain reaction (RT-PCR). Results Low amounts of quinoa (equivalent to 100 g/d of human intake) effectively controlled the weight of rats fed a high-fat diet. In addition, quinoa effectively inhibited the increase in hepatic TG and TC levels, mitigated pathological injury, promoted the increase in SOD and GSH-Px activities, and decreased MDA levels. Nontarget metabolic profile analysis showed that quinoa regulated lipid metabolites in the circulation system and liver such as LysoPC and PC. RNA-Seq and RT-PCR verification revealed that a high amount of quinoa more effectively upregulated genes related to lipid metabolism [Apoa (apolipoprotein)5, Apoa4, Apoc2] and downregulated genes related to the immune response [lrf (interferon regulatory factor)5, Tlr6 (Toll-like receptor), Tlr10, Tlr11, Tlr12]. Conclusion Quinoa effectively prevented NAFLD by controlling body weight, mitigating oxidative stress, and regulating the lipid metabolic profile and the expression of genes related to lipid metabolism and the immune response. Supplementary Information The online version contains supplementary material available at 10.1186/s12986-021-00631-7.
Collapse
Affiliation(s)
- Chenwei Song
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Wei Lv
- National Semi-Arid Agriculture Engineering Technology Research Center, Shijiazhuang, 050051, Hebei, China
| | - Yahui Li
- Center for Food Evaluation, State Administration for Market Regulation, Beijing, 100070, China
| | - Pan Nie
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jun Lu
- Shanghai Center for Plant Stress Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai, 201602, China
| | - Yanlou Geng
- National Semi-Arid Agriculture Engineering Technology Research Center, Shijiazhuang, 050051, Hebei, China.
| | - Zhang Heng
- Shanghai Center for Plant Stress Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai, 201602, China.
| | - Lihua Song
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
7
|
Association between Serum Uric Acid and Non-Alcoholic Fatty Liver Disease according to Different Menstrual Status Groups. Can J Gastroenterol Hepatol 2019; 2019:2763093. [PMID: 31871925 PMCID: PMC6906828 DOI: 10.1155/2019/2763093] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 10/22/2019] [Accepted: 10/28/2019] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE The present study aimed to explore the association between SUA and NAFLD in women with different menstrual statuses. METHODS A total of 6043 women were selected from the Jidong and Kailuan communities for inclusion in the present study. The SUA levels of participants were divided into quartiles. NAFLD was determined by abdominal ultrasonography. Data from laboratory tests and clinical examination were collected, and basic information was obtained from standardized questionnaires. The menstrual status was stratified into menstrual period, menopause transition period, and postmenopause. Multivariate logistic regression models were used to determine the relationship between menstrual status, SUA, and NAFLD. RESULTS The levels of SUA in subjects with NAFLD in the menstrual period, menopause transition period, and postmenopause were 268.0 ± 71.1, 265.6 ± 67.8, and 286.7 ± 75.8 (mmol/L), respectively, and were higher than those in subjects without NAFLD. The adjusted odds ratios (ORs) with 95% confidence interval (CI) for NAFLD among participants in the menopause transition period and postmenopausal period were 1.10 (0.89-1.37) and 1.28 (1.04-1.58), respectively, compared with the menstrual period women. Compared to the lowest quartile of SUA, the adjusted ORs with 95% CI of the highest quartile for NAFLD were 2.24 (1.69-2.99) for females in the menstrual period, 1.92 (1.10-3.37) for females in the menopause transition period, and 1.47 (1.06-2.03) for females in postmenopause. CONCLUSIONS Menstrual status was significantly correlated with NAFLD. High levels of SUA were associated with NAFLD in females during the three menstrual periods.
Collapse
|
8
|
Zhou YJ, Chang YN, You JQ, Li SZ, Zhuang W, Cao CJ. Cold-pressed Canola Oil Reduces Hepatic Steatosis by Modulating Oxidative Stress and Lipid Metabolism in KM Mice Compared with Refined Bleached Deodorized Canola Oil. J Food Sci 2019; 84:1900-1908. [PMID: 31183867 DOI: 10.1111/1750-3841.14504] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 01/15/2019] [Accepted: 02/14/2019] [Indexed: 12/15/2022]
Abstract
The quality of canola oil is affected by different extraction methods. The effect of cold-pressed canola oil (CPCO) diet and traditional refined bleached deodorized canola oil (RBDCO) diet on lipid accumulation and hepatic steatosis in mice were investigated. The body weight, peroxisome proliferator-activated receptor-α concentration, serum lipid profile, insulin sensitivity, and oxidative stress were increased in mice fed with CPCO diet, which had higher unsaturated fatty acid, tocopherols, phytosterols, and phospholipids but lower saturated fatty acid than RBDCO, after 12 weeks,. Moreover, CPCO significantly increased tocopherols and phytosterols content in liver and reduced liver cholesterol contents and lipid vacuoles accumulation than RBDCO. Also, serum proinflammatory cytokines, 3-hydroxy-3-methylglutary coenzyme A reductase expression level, lipogenic enzymes, and transcriptional factors such as sterol regulatory element-binding proteins 1c, acetyl-CoA carboxylase, and fatty acid synthase in the liver were also markedly downregulated from CPCO diet mice. Overall, CPCO can reduce lipid accumulation and hepatic steatosis by regulating oxidative stress and lipid metabolism in Kun Ming mice compared with RBDCO. PRACTICAL APPLICATION: The results suggested that more bioactive components were contained in cold-pressed canola oil (CPCO) rather than refined bleached deodorized canola oil (RBDCO). CPCO could lower the risk of obesity and hyperlipidemia, reduce lipid accumulation, and prevent hepatic steatosis. It could be considered as a kind of better edible oil than RBDCO.
Collapse
Affiliation(s)
- Ying-Jun Zhou
- The State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China Univ. of Science and Technology, Xuhui District, Shanghai, China
| | - Ya-Ning Chang
- The State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China Univ. of Science and Technology, Xuhui District, Shanghai, China
| | - Jia-Qi You
- The State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China Univ. of Science and Technology, Xuhui District, Shanghai, China
| | - Sui-Zi Li
- The State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China Univ. of Science and Technology, Xuhui District, Shanghai, China
| | - Wei Zhuang
- The State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China Univ. of Science and Technology, Xuhui District, Shanghai, China
| | - Cheng-Jia Cao
- The State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China Univ. of Science and Technology, Xuhui District, Shanghai, China
| |
Collapse
|
9
|
Liang H, Xie X, Song X, Huang M, Su T, Chang X, Liang B, Huang D. Orphan nuclear receptor NR4A1 suppresses hyperhomocysteinemia-induced hepatic steatosis in vitro and in vivo. FEBS Lett 2019; 593:1061-1071. [PMID: 30973961 DOI: 10.1002/1873-3468.13384] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 03/20/2019] [Accepted: 04/08/2019] [Indexed: 02/05/2023]
Abstract
Homocysteine (Hcy) is associated with nonalcoholic fatty liver disease (NAFLD). orphan nuclear receptor subfamily 4 group A member 1 (NR4A1) is involved in hepatic lipid metabolism. However, the potential role of NR4A1 in Hcy-associated NAFLD remains elusive. We aimed to elucidate the regulation of NR4A1 and its significance in Hcy-induced NAFLD. Hcy induced steatosis and elevated the expression of CD36 and FATP2 in HepG2 cells. Furthermore, Hcy enhanced p300 and decreased HDAC7 recruitment to the NR4A1 promoter, resulting in histone H3K27 hyperacetylation and NR4A1 upregulation. Moreover, NR4A1 depletion not only mimicked but also exaggerated the effects of Hcy on steatosis, whereas NR4A1 agonist Cytosporone B (CsnB) blocked Hcy-induced steatosis. In hyperhomocysteinemia (HHcy) mice, CsnB attenuated HHcy-induced hepatic steatosis. Thus, Hcy transiently and rapidly induces NR4A1 expression to reduce Hcy-induced steatosis.
Collapse
Affiliation(s)
- Hongjin Liang
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, China
- Department of Rheumatology, the First Affiliated Hospital, Shantou University Medical College, China
| | - Xina Xie
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, China
- Health Science Center, Institute of Translational Medicine, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, China
| | - Xuhong Song
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, China
| | - Meihui Huang
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, China
- Department of Pathology and Central Laboratory, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, China
| | - Ting Su
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, China
| | - Xiaolan Chang
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, China
| | - Bin Liang
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, China
| | - Dongyang Huang
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, China
| |
Collapse
|
10
|
Golabi P, Paik J, Hwang JP, Wang S, Lee HM, Younossi ZM. Prevalence and outcomes of non-alcoholic fatty liver disease (NAFLD) among Asian American adults in the United States. Liver Int 2019; 39:748-757. [PMID: 30597715 DOI: 10.1111/liv.14038] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/21/2018] [Accepted: 12/25/2018] [Indexed: 02/13/2023]
Abstract
BACKGROUND & AIMS Non-alcoholic fatty liver disease (NAFLD) affects about 25% of the general population worldwide. Although epidemiology of NAFLD is well studied in the United States, there is paucity of data for the Asian Americans. Our aim was to assess the prevalence and risk factors for NAFLD among Asian Americans. METHODS We utilized NHANES data for 2011-2016. We defined NAFLD using recently derived US-FLI. Relative risks (RRs) and population attributable fractions (PAFs) of metabolic components on atherosclerotic cardiovascular disease (ASCVD) and advanced fibrosis were calculated for Asian Americans, and these rates were compared to non-Hispanic whites. RESULTS NAFLD prevalence was 18.3% among Asian Americans and 28.4% among non-Hispanic whites. Asian Americans with NAFLD had lower BMI and waist circumference than non-Hispanic whites with NAFLD and were less likely to have metabolic syndrome, cardiovascular disease (CVD), chronic obstructive pulmonary disease, cancer and incident ASCVD (P < 0.05). Hyperlipidaemia had the highest attributable fraction (76.6%) for risk of ASCVD among Asian Americans with NAFLD, followed by diabetes (24.0%), current smoking (9.2%), and obesity (3.7%). Advanced fibrosis in Asian American with NAFLD was independently associated with presence of type 2 diabetes (RR = 2.70, 95% CI: 1.00-7.27). CONCLUSIONS Asian Americans have lower prevalence of NAFLD than non-Hispanic whites. However, Asian Americans with NAFLD have similar risk factors for advanced fibrosis and ASCVD than non-Hispanic Whites.
Collapse
Affiliation(s)
- Pegah Golabi
- Betty and Guy Beatty Center for Integrated Research, Inova Health System, Falls Church, Virginia
| | - James Paik
- Betty and Guy Beatty Center for Integrated Research, Inova Health System, Falls Church, Virginia
| | - Jessica P Hwang
- Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Su Wang
- Center for Asian Health, Saint Barnabas Medical Center, Florham Park, New Jersey
| | - Hannah M Lee
- Division of Gastroenterology, Hepatology, and Nutrition, Virginia Commonwealth University, Richmond, Virginia
| | - Zobair M Younossi
- Betty and Guy Beatty Center for Integrated Research, Inova Health System, Falls Church, Virginia.,Center for Liver Disease and Department of Medicine, Inova Fairfax Medical Campus, Falls Church, Virginia
| |
Collapse
|
11
|
Zhu SY, Jiang N, Yang J, Tu J, Zhou Y, Xiao X, Dong Y. Silybum marianum oil attenuates hepatic steatosis and oxidative stress in high fat diet-fed mice. Biomed Pharmacother 2018; 100:191-197. [PMID: 29428667 DOI: 10.1016/j.biopha.2018.01.144] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 01/08/2018] [Accepted: 01/28/2018] [Indexed: 02/08/2023] Open
Abstract
In the present study, the effects of Silybum marianum oil (SMO) on hepatic steatosis and oxidative stress were investigated during the development of nonalcoholic fatty liver disease (NAFLD) in high fat diet (HFD)-fed mice. The results showed that body weight, fat mass, and serum biochemical parameters such as triglyceride, free fatty acid, glucose and insulin were reduced by SMO treatment. Meanwhile, SMO decreased the histological injury of liver and the levels of hepatic triglyceride, cholesterol and free fatty acid in HFD-fed mice. SMO administration elevated the activities of superoxide dismutase (SOD) and catalase (CAT) and reduced the level of malondialdehyde (MDA) in the liver. Enzyme linked immunosorbent assay showed that SMO significantly decreased the levels of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) in HFD mice. Furthermore, the mRNA levels of sterol regulatory element binding protein 1c (SREBP-1c), fatty acid synthase (FAS) and liver X receptor α (LXRα) were lower, but peroxisome proliferator-activated receptor α (PPARα) was higher in mice treated with SMO compared with the HFD group. The results indicated that SMO could play a certain protective role against HFD-induced NAFLD, and the protective effects might be associated with attenuating lipid accumulation, oxidative stress and inflammation, improving lipid metabolism.
Collapse
Affiliation(s)
- Shu Yun Zhu
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, China; Jiangsu Hengshun Group Co., Ltd., Zhenjiang 212000, China.
| | - Ning Jiang
- Institute of Vegetables, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Jing Yang
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Jie Tu
- College of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212018, China
| | - Yue Zhou
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Xiang Xiao
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Ying Dong
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
12
|
Kim H, Lee KW, Lee K, Seo S, Park MY, Ahn SW, Hong SK, Yoon KC, Kim HS, Choi Y, Lee HW, Yi NJ, Suh KS. Effect of PNPLA3 I148M polymorphism on histologically proven non-alcoholic fatty liver disease in liver transplant recipients. Hepatol Res 2018; 48:E162-E171. [PMID: 28718984 DOI: 10.1111/hepr.12940] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 07/10/2017] [Accepted: 07/13/2017] [Indexed: 12/13/2022]
Abstract
AIM PNPLA3 I148M polymorphism (rs738409 C>G) is the most important and best-known polymorphism for non-alcoholic fatty liver disease (NAFLD). However, little is known about the effect of this polymorphism on NAFLD after liver transplantation (LT). We aimed to evaluate the association between this polymorphism and post-LT NAFLD. METHODS We designed a prospective case-control study. Among adult recipients who underwent LT between April 2014 and October 2015, those whose whole blood was preoperatively collected for genotyping in both recipients and coupled donors and those who underwent protocol biopsy at 1 year post-LT were enrolled. RESULTS A total of 32 recipients were enrolled. Histologically proven steatosis (≥5%) was present in 28.1% of patients at a mean time of 12.7 ± 2.0 months after LT. Moderate and more severe steatosis (≥33%) was present in 9.4%. One year after LT, steatosis was present in 50.0% of homozygous recipients with the rs738409-G allele. It was present in 27.3% of heterozygous recipients with the rs738409-G allele, and in 9.1% (P = 0.041) of recipients with rs738409-CC. The genotype of the donor was not significantly (P = 0.647) associated with post-LT NAFLD. When both recipient and coupled donor showed heterogeneous or homozygous genotype of the rs738409-G allele, there was significantly more post-LT NAFLD compared to that in others (47.1% vs. 6.7%; P = 0.018). In univariate and multivariate analyses, only the presence of the rs738409-G risk allele in both donor and recipient was a significant risk factor for post-LT NALFD (relative risk, 26.95; P = 0.048). CONCLUSIONS PNPLA3 I148M polymorphism can significantly affect histologically proven NAFLD at 1 year post-LT.
Collapse
Affiliation(s)
- Hyeyoung Kim
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Kwang-Woong Lee
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Kyoungbun Lee
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - Sooin Seo
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Min-Young Park
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Sung Woo Ahn
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Suk Kyun Hong
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Kyung Chul Yoon
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Hyo-Sin Kim
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - YoungRok Choi
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Hae Won Lee
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Nam-Joon Yi
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Kyung-Suk Suh
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
13
|
Serra-Planas E, Aguilera E, Castro L, Rodríguez R, Salinas I, Lucas A, Joaquín C, Puig R, Mauricio D, Puig-Domingo M. Low prevalence of non-alcoholic fatty liver disease in patients with type 1 diabetes is associated with decreased subclinical cardiovascular disease. J Diabetes 2017; 9:1065-1072. [PMID: 28220621 DOI: 10.1111/1753-0407.12539] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 01/10/2017] [Accepted: 02/16/2017] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) has been proposed as an independent cardiovascular risk factor. The present study evaluated the prevalence of NAFLD in a cohort of type 1 diabetic (T1D) patients and its potential relationship with subclinical cardiovascular disease (CVD). METHODS One hundred T1D patients (mean [±SD] age 39.4 ± 7.8 years, disease duration 21.7 ± 8.6 years) were included in the present cross-sectional study. All subjects underwent abdominal ultrasonography for detection of NAFLD, carotid ultrasonography to measure the carotid intima-media thickness (CIMT) and atheroma plaques, and cardiac tomography for evaluation of the coronary artery calcium score (CACS). RESULTS Of the study cohort, 12% had NAFLD and 23% had a CACS >0. The T1D subjects with NAFLD had a greater CIMT than those without NAFLD (0.65 ± 0.17 vs 0.55 ± 0.14 mm; P = 0.029), but there were no significant differences between the two groups with regard to CACS, glycemic control, or the presence of carotid plaques. Patients with high liver enzyme concentrations (>20 U/L) had a higher CIMT (0.60 ± 0.16 vs 0.54 ± 0.13; P = 0.04) and there was a higher proportion of altered CACS (17 [73.9%] vs 6 [26.1%]; P = 0.001) and detection of carotid plaques (10 [76.9%] vs 3 [23.1%]; P = 0.014) in this group. CONCLUSIONS A low prevalence of NAFLD was found in the T1D cohort that was associated globally with a low proportion of abnormal CVD imaging markers, although these imaging parameters were worse in subjects in whom NAFLD was detected.
Collapse
Affiliation(s)
- Enric Serra-Planas
- Department of Endocrinology and Nutrition, Health Sciences Research Institute and University Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Eva Aguilera
- Department of Endocrinology and Nutrition, Health Sciences Research Institute and University Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
- Department of Endocrinology and Nutrition, CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Health Sciences Research Institute and University Hospital Germans Trias i Pujol, Badalona, Spain
| | - Laura Castro
- Service of Radiology, Institute of Research and Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Raúl Rodríguez
- Service of Radiology, Institute of Research and Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Isabel Salinas
- Department of Endocrinology and Nutrition, Health Sciences Research Institute and University Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Anna Lucas
- Department of Endocrinology and Nutrition, Health Sciences Research Institute and University Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
- Department of Endocrinology and Nutrition, CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Health Sciences Research Institute and University Hospital Germans Trias i Pujol, Badalona, Spain
| | - Clara Joaquín
- Department of Endocrinology and Nutrition, Health Sciences Research Institute and University Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Rocio Puig
- Department of Endocrinology and Nutrition, Health Sciences Research Institute and University Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Dídac Mauricio
- Department of Endocrinology and Nutrition, Health Sciences Research Institute and University Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
- Department of Endocrinology and Nutrition, CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Health Sciences Research Institute and University Hospital Germans Trias i Pujol, Badalona, Spain
| | - Manel Puig-Domingo
- Department of Endocrinology and Nutrition, Health Sciences Research Institute and University Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
- Department of Endocrinology and Nutrition, CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Health Sciences Research Institute and University Hospital Germans Trias i Pujol, Badalona, Spain
| |
Collapse
|
14
|
Geng C, Xu H, Zhang Y, Gao Y, Li M, Liu X, Gao M, Wang X, Liu X, Fang F, Chang Y. Retinoic acid ameliorates high-fat diet-induced liver steatosis through sirt1. SCIENCE CHINA-LIFE SCIENCES 2017; 60:1234-1241. [PMID: 28667519 DOI: 10.1007/s11427-016-9027-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 03/17/2017] [Indexed: 12/17/2022]
Abstract
In this study, treatment of C57BL/6J (wild type, WT) mice fed a high-fat diet (HFD) with retinoic acid (RA) decreased body weight and subcutaneous and visceral fat content, reversed the apparent hepatosteatosis, and reduced hepatic intracellular triglyceride and serum alanine transaminase (ALT) and aspartate aminotransferase (AST) concentrations. Moreover, RA treatment improved glucose tolerance and insulin sensitivity in WT mice fed a HFD. However, these RA-induced effects in WT mice fed a HFD were alleviated in liver specific Sirtuin 1 (Sirt1) deficient (LKO) mice fed a HFD. Furthermore, RA also could not improve glucose tolerance and insulin sensitivity in LKO mice fed a HFD. The mechanism studies indicated that RA indeed increased the expression of hepatic Sirt1 and superoxide dismutase 2 (Sod2), and inhibited the expression of sterol regulatory element binding protein 1c (Srebp-1c) in WT mice in vivo and in vitro. RA decreased mitochondrial reactive oxygen species (ROS) production in WT primary hepatocytes and increased mitochondrial DNA (mtDNA) copy number in WT mice liver. However, these RA-mediated molecular effects were also abolished in the liver and primary hepatocytes from LKO mice. In summary, RA protected against HFD-induced hepatosteatosis by decreasing Srebp-1c expression and improving antioxidant capacity through a Sirt1-mediated mechanism.
Collapse
Affiliation(s)
- Chao Geng
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Haifeng Xu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Interventional Therapy, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Yinliang Zhang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Yong Gao
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Meixia Li
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xiaoyan Liu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Mingyue Gao
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Xiaojuan Wang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Xiaojun Liu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine Peking Union Medical College, Beijing, 100005, China.
| | - Fude Fang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine Peking Union Medical College, Beijing, 100005, China.
| | - Yongsheng Chang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine Peking Union Medical College, Beijing, 100005, China.
| |
Collapse
|
15
|
Watanabe S, Takahashi T, Ogawa H, Uehara H, Tsunematsu T, Baba H, Morimoto Y, Tsuneyama K. Daily Coffee Intake Inhibits Pancreatic Beta Cell Damage and Nonalcoholic Steatohepatitis in a Mouse Model of Spontaneous Metabolic Syndrome, Tsumura-Suzuki Obese Diabetic Mice. Metab Syndr Relat Disord 2017; 15:170-177. [PMID: 28358620 DOI: 10.1089/met.2016.0114] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Metabolic syndrome is one of the most important health issues worldwide. Obesity causes insulin resistance, hyperlipidemia, diabetes, and various diseases throughout the body. The liver phenotype, which is called nonalcoholic steatohepatitis (NASH), frequently progresses to hepatocellular carcinoma. We recently established a new animal model, Tsumura-Suzuki obese diabetic (TSOD) mice, which spontaneously exhibit obesity, diabetes, hyperlipidemia, and NASH with liver nodules. METHODS We examined the effects of coffee intake on various conditions of the metabolic syndrome using TSOD mice. The daily volume of coffee administered was limited so that it reflected the appropriate quantities consumed in humans. To clarify the effects of the specific components, animals were divided into two coffee-intake groups that included with and without caffeine. RESULTS Coffee intake did not significantly affect obesity and hyperlipidemia in TSOD mice. In contrast, coffee intake caused various degrees of improvement in the pancreatic beta cell damage and steatohepatitis with liver carcinogenesis. Most of the effects were believed to be caused by a synergistic effect of caffeine with other components such as polyphenols. However, the antifibrotic effects of coffee appeared to be due to the polyphenols rather than the caffeine. CONCLUSIONS A daily habit of drinking coffee could possibly play a role in the prevention of metabolic syndrome.
Collapse
Affiliation(s)
- Syunsuke Watanabe
- 1 Department of Pathology and Laboratory Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School , Tokushima, Japan
| | - Tetsuyuki Takahashi
- 2 Department of Anatomy and Cell Biology, Faculty of Pharmacy, Research Institute of Pharmaceutical Science, Musashino University , Nishitokyo, Japan
| | - Hirohisa Ogawa
- 1 Department of Pathology and Laboratory Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School , Tokushima, Japan
| | - Hisanori Uehara
- 1 Department of Pathology and Laboratory Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School , Tokushima, Japan
| | - Takaaki Tsunematsu
- 1 Department of Pathology and Laboratory Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School , Tokushima, Japan
| | - Hayato Baba
- 1 Department of Pathology and Laboratory Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School , Tokushima, Japan
| | - Yuki Morimoto
- 1 Department of Pathology and Laboratory Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School , Tokushima, Japan
| | - Koichi Tsuneyama
- 1 Department of Pathology and Laboratory Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School , Tokushima, Japan
| |
Collapse
|
16
|
Chardonnay Grape Seed Flour Ameliorates Hepatic Steatosis and Insulin Resistance via Altered Hepatic Gene Expression for Oxidative Stress, Inflammation, and Lipid and Ceramide Synthesis in Diet-Induced Obese Mice. PLoS One 2016; 11:e0167680. [PMID: 27977712 PMCID: PMC5157984 DOI: 10.1371/journal.pone.0167680] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 11/18/2016] [Indexed: 01/04/2023] Open
Abstract
To identify differentially expressed hepatic genes contributing to the improvement of high-fat (HF) diet-induced hepatic steatosis and insulin resistance following supplementation of partially defatted flavonoid-rich Chardonnay grape seed flour (ChrSd), diet-induced obese (DIO) mice were fed HF diets containing either ChrSd or microcrystalline cellulose (MCC, control) for 5 weeks. The 2-h insulin area under the curve was significantly lowered by ChrSd, indicating that ChrSd improved insulin sensitivity. ChrSd intake also significantly reduced body weight gain, liver and adipose tissue weight, hepatic lipid content, and plasma low-density lipoprotein (LDL)-cholesterol, despite a significant increase in food intake. Exon microarray analysis of hepatic gene expression revealed down-regulation of genes related to triglyceride and ceramide synthesis, immune response, oxidative stress, and inflammation and upregulation of genes related to fatty acid oxidation, cholesterol, and bile acid synthesis. In conclusion, the effects of ChrSd supplementation in a HF diet on weight gain, insulin resistance, and progression of hepatic steatosis in DIO mice were associated with modulation of hepatic genes related to oxidative stress, inflammation, ceramide synthesis, and lipid and cholesterol metabolism.
Collapse
|
17
|
Manna P, Jain SK. Obesity, Oxidative Stress, Adipose Tissue Dysfunction, and the Associated Health Risks: Causes and Therapeutic Strategies. Metab Syndr Relat Disord 2016; 13:423-44. [PMID: 26569333 DOI: 10.1089/met.2015.0095] [Citation(s) in RCA: 654] [Impact Index Per Article: 72.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Obesity is gaining acceptance as a serious primary health burden that impairs the quality of life because of its associated complications, including diabetes, cardiovascular diseases, cancer, asthma, sleep disorders, hepatic dysfunction, renal dysfunction, and infertility. It is a complex metabolic disorder with a multifactorial origin. Growing evidence suggests that oxidative stress plays a role as the critical factor linking obesity with its associated complications. Obesity per se can induce systemic oxidative stress through various biochemical mechanisms, such as superoxide generation from NADPH oxidases, oxidative phosphorylation, glyceraldehyde auto-oxidation, protein kinase C activation, and polyol and hexosamine pathways. Other factors that also contribute to oxidative stress in obesity include hyperleptinemia, low antioxidant defense, chronic inflammation, and postprandial reactive oxygen species generation. In addition, recent studies suggest that adipose tissue plays a critical role in regulating the pathophysiological mechanisms of obesity and its related co-morbidities. To establish an adequate platform for the prevention of obesity and its associated health risks, understanding the factors that contribute to the cause of obesity is necessary. The most current list of obesity determinants includes genetic factors, dietary intake, physical activity, environmental and socioeconomic factors, eating disorders, and societal influences. On the basis of the currently identified predominant determinants of obesity, a broad range of strategies have been recommended to reduce the prevalence of obesity, such as regular physical activity, ad libitum food intake limiting to certain micronutrients, increased dietary intake of fruits and vegetables, and meal replacements. This review aims to highlight recent findings regarding the role of oxidative stress in the pathogenesis of obesity and its associated risk factors, the role of dysfunctional adipose tissue in development of these risk factors, and potential strategies to regulate body weight loss/gain for better health benefits.
Collapse
Affiliation(s)
- Prasenjit Manna
- Department of Pediatrics, LSU Health Sciences Center , Shreveport, Louisiana
| | - Sushil K Jain
- Department of Pediatrics, LSU Health Sciences Center , Shreveport, Louisiana
| |
Collapse
|
18
|
Geng C, Zhang Y, Gao Y, Tao W, Zhang H, Liu X, Fang F, Chang Y. Mst1 regulates hepatic lipid metabolism by inhibiting Sirt1 ubiquitination in mice. Biochem Biophys Res Commun 2016; 471:444-9. [PMID: 26903296 DOI: 10.1016/j.bbrc.2016.02.059] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 02/15/2016] [Indexed: 12/11/2022]
Abstract
Previous study showed mammalian Ste20-like kinase (Mst1) may serve as target for the development of new therapies for diabetes. However, the function of Mst1 involved in liver lipid metabolism has remained elusive. In this study, we report that the liver of Mst1 knockout (Mst1(-/-)) mice showed more severe liver metabolic damage under fasting and high-fat diet than that of control mice. And fasting induced hepatic Mst1 expression. Mst1 overexpression inhibited Srebp-1c expression and increased the expression of antioxidant genes in primary hepatocytes. We also found that fasting-induced expression of hepatic Sirt1 was attenuated in Mst1(-/-) mice. Mst1 overexpression promoted Sirt1 expression, probably due to inhibiting Sirt1 ubiquitination. In summary, our study suggests that Mst1 regulates hepatic lipid metabolism by inhibiting Sirt1 ubiquitination in mice.
Collapse
Affiliation(s)
- Chao Geng
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences & School of Basic Medicine Peking Union Medical College, #5 Dongdansantiao, Beijing 100005, China
| | - Yinliang Zhang
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences & School of Basic Medicine Peking Union Medical College, #5 Dongdansantiao, Beijing 100005, China
| | - Yong Gao
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences & School of Basic Medicine Peking Union Medical College, #5 Dongdansantiao, Beijing 100005, China
| | - Wufan Tao
- State Key Laboratory of Genetic Engineering and Institute of Developmental Biology, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Huabing Zhang
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei 230032, China
| | - Xiaojun Liu
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences & School of Basic Medicine Peking Union Medical College, #5 Dongdansantiao, Beijing 100005, China.
| | - Fude Fang
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences & School of Basic Medicine Peking Union Medical College, #5 Dongdansantiao, Beijing 100005, China.
| | - Yongsheng Chang
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences & School of Basic Medicine Peking Union Medical College, #5 Dongdansantiao, Beijing 100005, China.
| |
Collapse
|
19
|
|
20
|
Liver cirrhosis mortality, alcohol consumption and tobacco consumption over a 62 year period in a high alcohol consumption country: a trend analysis. BMC Res Notes 2015; 8:822. [PMID: 26708239 PMCID: PMC4691532 DOI: 10.1186/s13104-015-1808-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 12/14/2015] [Indexed: 12/27/2022] Open
Abstract
Background The relationship between alcohol consumption and liver cirrhosis mortality has been revealed by data from several different countries. However, the impact of tobacco smoking on liver cirrhosis has not been considered. The aim of this study was to estimate trends in liver cirrhosis mortality and alcohol and tobacco consumption from 1952 to 2013 as well as more recent trends in substance use disorder treatments and hospital treatments of liver diseases in Germany. Methods Data from the National Statistics Office were used. Liver cirrhosis was diagnosed according to the International Classification of Diseases (ICD-6 to ICD-10). Alcohol beverages and tobacco products were estimated according to tax or governmental data. Substance use disorder treatment and hospital treatment data were used. Trends were calculated using Joinpoint regression analyses. Results Liver cirrhosis mortality among men increased annually by 8.4 % from 1952 to 1960 and increased annually by 2.8 % from 1961 to 1976. From 1976 to 1982, liver cirrhosis mortality decreased annually by 4.8 %, from 1982 to 2013 liver cirrhosis mortality decreased annually by 1.2 %. Among females, liver cirrhosis mortality increased annually by 8.9 % from 1952 to 1959 and by 4.3 % from 1959 to 1968, but then decreased 1.0 % annually from 1968 to 1995. After 1995, liver cirrhosis mortality decreased 1.9 % annually through 2013. These reductions in liver cirrhosis mortality were accompanied by decreases in alcohol consumption beginning in 1976. These findings were also accompanied by decreases in the consumption of cigarette equivalents since 1971. Meanwhile, the number of substance use disorder treatments and hospital treatments of liver diseases increased. Conclusions The decrease in liver cirrhosis mortality may have been caused by a decrease in alcohol drinking and tobacco smoking. Smoking may have exerted indirect effects via alcohol consumption as well as direct effects. These trends existed despite largely missing preventive efforts to reduce alcohol consumption and tobacco smoking. Increases in educational attainment in the general population may have contributed to the reductions in alcohol and tobacco consumption. Convincing evidence that the increased provision of substance use disorder treatment significantly contributed to the decrease of liver cirrhosis was not found. Electronic supplementary material The online version of this article (doi:10.1186/s13104-015-1808-2) contains supplementary material, which is available to authorized users.
Collapse
|
21
|
Lambert JE, Parnell JA, Eksteen B, Raman M, Bomhof MR, Rioux KP, Madsen KL, Reimer RA. Gut microbiota manipulation with prebiotics in patients with non-alcoholic fatty liver disease: a randomized controlled trial protocol. BMC Gastroenterol 2015; 15:169. [PMID: 26635079 PMCID: PMC4669628 DOI: 10.1186/s12876-015-0400-5] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 11/25/2015] [Indexed: 02/08/2023] Open
Abstract
Background Evidence for the role of the gut microbiome in the pathogenesis of non-alcoholic fatty liver disease (NAFLD) is emerging. Strategies to manipulate the gut microbiota towards a healthier community structure are actively being investigated. Based on their ability to favorably modulate the gut microbiota, prebiotics may provide an inexpensive yet effective dietary treatment for NAFLD. Additionally, prebiotics have established benefits for glucose control and potentially weight control, both advantageous in managing fatty liver disease. Our objective is to evaluate the effects of prebiotic supplementation, adjunct to those achieved with diet-induced weight loss, on heptic injury and liver fat, the gut microbiota, inflammation, glucose tolerance, and satiety in patients with NAFLD. Methods/design In a double blind, placebo controlled, parallel group study, adults (BMI ≥25) with confirmed NAFLD will be randomized to either a 16 g/d prebiotic supplemented group or isocaloric placebo group for 24 weeks (n = 30/group). All participants will receive individualized dietary counseling sessions with a registered dietitian to achieve 10 % weight loss. Primary outcome measures include change in hepatic injury (fibrosis and inflammation) and liver fat. Secondary outcomes include change in body composition, appetite and dietary adherence, glycemic and insulinemic responses and inflammatory cytokines. Mechanisms related to prebiotic-induced changes in gut microbiota (shot-gun sequencing) and their metabolic by-products (volatile organic compounds) and de novo lipogenesis (using deuterium incorporation) will also be investigated. Discussion There are currently no medications or surgical procedures approved for the treatment of NAFLD and weight loss via lifestyle modification remains the cornerstone of current care recommendations. Given that prebiotics target multiple metabolic impairments associated with NAFLD, investigating their ability to modulate the gut microbiota and hepatic health in patients with NAFLD is warranted. Trial registration ClinicalTrials.gov (NCT02568605) Registered 30 September 2015
Collapse
Affiliation(s)
- Jennifer E Lambert
- Faculty of Kinesiology, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada.
| | - Jill A Parnell
- Health and Physical Education, Mount Royal University, 4825 Mount Royal Gate SW, Calgary, AB, T3E 6K6, Canada.
| | - Bertus Eksteen
- Snyder Institute for Chronic Diseases, Health Research and Innovation Center, University of Calgary, 3280 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada. .,Division of Gastroenterology and Hepatology, Department of Medicine, University of Calgary, 3280 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada.
| | - Maitreyi Raman
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Calgary, 3280 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada.
| | - Marc R Bomhof
- Faculty of Kinesiology, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada.
| | - Kevin P Rioux
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Calgary, 3280 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada. .,Department of Microbiology and Infectious Diseases, University of Calgary, 1863 Health Sciences Centre, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada.
| | - Karen L Madsen
- Division of Gastroenterology, Centre of Excellence for Gastrointestinal Inflammation and Immunity Research, 7-142 Katz Group-Rexall Centre, University of Alberta, Edmonton, AB, T6G 2C2, Canada.
| | - Raylene A Reimer
- Faculty of Kinesiology, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada. .,Department of Biochemistry & Molecular Biology, Cumming School of Medicine, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada.
| |
Collapse
|
22
|
Abstract
Associated with the obesity epidemic, non-alcoholic fatty liver disease (NAFLD) has become the leading liver disease in North America. Approximately 30 % of patients with NAFLD may develop non-alcoholic steatohepatitis (NASH) that can lead to cirrhosis and hepatocellular carcinoma (HCC). Frequently animal models are used to help identify underlying factors contributing to NAFLD including insulin resistance, dysregulated lipid metabolism and mitochondrial stress. However, studying the inflammatory, progressive nature of NASH in the context of obesity has proven to be a challenge in mice. Although the development of effective treatment strategies for NAFLD and NASH is gaining momentum, the field is hindered by a lack of a concise animal model that reflects the development of liver disease during obesity and the metabolic syndrome. Therefore, selecting an animal model to study NAFLD or NASH must be done carefully to ensure the optimal application. The most widely used animal models have been reviewed highlighting their advantages and disadvantages to studying NAFLD and NASH specifically in the context of obesity.
Collapse
|
23
|
Recent insights on the role of cholesterol in non-alcoholic fatty liver disease. Biochim Biophys Acta Mol Basis Dis 2015; 1852:1765-78. [DOI: 10.1016/j.bbadis.2015.05.015] [Citation(s) in RCA: 175] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 05/25/2015] [Accepted: 05/27/2015] [Indexed: 12/18/2022]
|
24
|
Pizarro M, Solís N, Quintero P, Barrera F, Cabrera D, Santiago PR, Arab JP, Padilla O, Roa JC, Moshage H, Wree A, Inzaugarat E, Feldstein AE, Fardella CE, Baudrand R, Riquelme A, Arrese M. Beneficial effects of mineralocorticoid receptor blockade in experimental non-alcoholic steatohepatitis. Liver Int 2015; 35:2129-38. [PMID: 25646700 PMCID: PMC4522413 DOI: 10.1111/liv.12794] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 01/28/2015] [Indexed: 12/14/2022]
Abstract
BACKGROUND Therapeutic options to treat Non-alcoholic steatohepatitis (NASH) are limited. Mineralocorticoid receptor (MR) activation could play a role in hepatic fibrogenesis and its modulation could be beneficial for NASH. AIM To investigate whether eplerenone, a specific MR antagonist, ameliorates liver damage in experimental NASH. METHODS C57bl6 mice were fed a choline-deficient and amino acid-defined (CDAA) diet for 22 weeks with or without eplerenone supplementation. Serum levels of aminotransferases and aldosterone were measured and hepatic steatosis, inflammation and fibrosis scored histologically. Hepatic triglyceride content (HTC) and hepatic mRNA levels of pro-inflammatory pro-fibrotic, oxidative stress-associated genes and of MR were also assessed. RESULTS CDAA diet effectively induced fibrotic NASH, and increased the hepatic expression of pro-inflammatory, pro-fibrotic and oxidative stress-associated genes. Hepatic MR mRNA levels significantly correlated with the expression of pro-inflammatory and pro-fibrotic genes and were significantly increased in hepatic stellate cells obtained from CDAA-fed animals. Eplerenone administration was associated to a reduction in histological steatosis and attenuation of liver fibrosis development, which was associated to a significant decrease in the expression of collagen-α1, collagen type III, alpha 1 and Matrix metalloproteinase-2. CONCLUSION The expression of MR correlates with inflammation and fibrosis development in experimental NASH. Specific MR blockade with eplerenone has hepatic anti-steatotic and anti-fibrotic effects. These data identify eplerenone as a potential novel therapy for NASH. Considering its safety and FDA-approved status, human studies are warranted.
Collapse
Affiliation(s)
- Margarita Pizarro
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Nancy Solís
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Pablo Quintero
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Francisco Barrera
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Daniel Cabrera
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile,Departamento de Ciencias Químico-Biológicas, Universidad Bernardo O'Higgins, Santiago, Chile
| | - Pamela Rojasde Santiago
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Juan Pablo Arab
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Oslando Padilla
- Departamento de Salud Pública, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan Carlos Roa
- Departamento de Patología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Han Moshage
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Alexander Wree
- Department of Pediatrics, University of California, San Diego, CA, USA
| | | | | | - Carlos E. Fardella
- Departamento de Endocrinología, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Rene Baudrand
- Departamento de Endocrinología, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Arnoldo Riquelme
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Marco Arrese
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile,Corresponding author: Marco Arrese, M.D. Department of Gastroenterology Escuela de Medicina Pontificia Universidad Católica de Chile Marcoleta #367 833-0024 Santiago CHILE Phone/Fax: 56-2-6397780,
| |
Collapse
|
25
|
Chen PJ, Cai SP, Huang C, Meng XM, Li J. Protein tyrosine phosphatase 1B (PTP1B): A key regulator and therapeutic target in liver diseases. Toxicology 2015; 337:10-20. [PMID: 26299811 DOI: 10.1016/j.tox.2015.08.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 08/12/2015] [Accepted: 08/15/2015] [Indexed: 12/11/2022]
Abstract
Phosphorylation of tyrosine residues within proteins, which is controlled by the reciprocal action of protein tyrosine kinases and protein tyrosine phosphatases, plays a key role in regulating almost all physiological responses. Therefore, it comes as no surprise that once the balance of tyrosine phosphorylation is disturbed, drastic effects can occur. Protein tyrosine phosphatase 1B (PTP1B), a classical non-transmembrane tyrosine phosphatase, is a pivotal regulator and promising drug target in type 2 diabetes and obesity. Recently it has received renewed attention in liver diseases and represents an intriguing opportunity as a drug target by modulating hepatocyte death and survival, hepatic lipogenesis and so on. Here, the multiple roles of PTP1B in liver diseases will be presented, with respect to liver regeneration, drug-induced liver disease, non-alcoholic fatty liver disease (NAFLD) and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Pei-Jie Chen
- School of Pharmacy, Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University (ILD-AMU), Anhui Medical University, Hefei 230032, China
| | - Shuang-Peng Cai
- School of Pharmacy, Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University (ILD-AMU), Anhui Medical University, Hefei 230032, China
| | - Cheng Huang
- School of Pharmacy, Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University (ILD-AMU), Anhui Medical University, Hefei 230032, China
| | - Xiao-Ming Meng
- School of Pharmacy, Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University (ILD-AMU), Anhui Medical University, Hefei 230032, China
| | - Jun Li
- School of Pharmacy, Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University (ILD-AMU), Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
26
|
LIN JIAYAO, ZHANG YU, WANG XINQING, WANG WENWEN. Lycium ruthenicum extract alleviates high-fat diet-induced nonalcoholic fatty liver disease via enhancing the AMPK signaling pathway. Mol Med Rep 2015; 12:3835-3840. [DOI: 10.3892/mmr.2015.3840] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 04/30/2015] [Indexed: 11/06/2022] Open
|
27
|
Canet MJ, Merrell MD, Hardwick RN, Bataille AM, Campion SN, Ferreira DW, Xanthakos SA, Manautou JE, A-Kader HH, Erickson RP, Cherrington NJ. Altered regulation of hepatic efflux transporters disrupts acetaminophen disposition in pediatric nonalcoholic steatohepatitis. Drug Metab Dispos 2015; 43:829-35. [PMID: 25788542 DOI: 10.1124/dmd.114.062703] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Accepted: 03/12/2015] [Indexed: 12/31/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease, representing a spectrum of liver pathologies that include simple hepatic steatosis and the more advanced nonalcoholic steatohepatitis (NASH). The current study was conducted to determine whether pediatric NASH also results in altered disposition of acetaminophen (APAP) and its two primary metabolites, APAP-sulfate and APAP-glucuronide. Pediatric patients with hepatic steatosis (n = 9) or NASH (n = 3) and healthy patients (n = 12) were recruited in a small pilot study design. All patients received a single 1000-mg dose of APAP. Blood and urine samples were collected at 1, 2, and 4 hours postdose, and APAP and APAP metabolites were determined by high-performance liquid chromatography. Moreover, human liver tissues from patients diagnosed with various stages of NAFLD were acquired from the Liver Tissue Cell Distribution System to investigate the regulation of the membrane transporters, multidrug resistance-associated protein 2 and 3 (MRP2 and MRP3, respectively). Patients with the more severe disease (i.e., NASH) had increased serum and urinary levels of APAP-glucuronide along with decreased serum levels of APAP-sulfate. Moreover, an induction of hepatic MRP3 and altered canalicular localization of the biliary efflux transporter, MRP2, describes the likely mechanism for the observed increase in plasma retention of APAP-glucuronide, whereas altered regulation of sulfur activation genes may explain decreased sulfonation activity in NASH. APAP-glucuronide and APAP-sulfate disposition is altered in NASH and is likely due to hepatic membrane transporter dysregulation as well as altered intracellular sulfur activation.
Collapse
Affiliation(s)
- Mark J Canet
- Departments of Pharmacology and Toxicology (M.J.C., M.D.M., R.N.H., N.J.C.) and Pediatrics (H.A.K., R.P.E.), University of Arizona, Tucson, Arizona; School of Pharmacy, University of Connecticut, Storrs, Connecticut (A.M.B., D.W.F., J.E.M.); Drug Safety Research and Development, Pfizer, Inc., New York, New York (S.N.C.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio (S.A.X.)
| | - Matthew D Merrell
- Departments of Pharmacology and Toxicology (M.J.C., M.D.M., R.N.H., N.J.C.) and Pediatrics (H.A.K., R.P.E.), University of Arizona, Tucson, Arizona; School of Pharmacy, University of Connecticut, Storrs, Connecticut (A.M.B., D.W.F., J.E.M.); Drug Safety Research and Development, Pfizer, Inc., New York, New York (S.N.C.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio (S.A.X.)
| | - Rhiannon N Hardwick
- Departments of Pharmacology and Toxicology (M.J.C., M.D.M., R.N.H., N.J.C.) and Pediatrics (H.A.K., R.P.E.), University of Arizona, Tucson, Arizona; School of Pharmacy, University of Connecticut, Storrs, Connecticut (A.M.B., D.W.F., J.E.M.); Drug Safety Research and Development, Pfizer, Inc., New York, New York (S.N.C.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio (S.A.X.)
| | - Amy M Bataille
- Departments of Pharmacology and Toxicology (M.J.C., M.D.M., R.N.H., N.J.C.) and Pediatrics (H.A.K., R.P.E.), University of Arizona, Tucson, Arizona; School of Pharmacy, University of Connecticut, Storrs, Connecticut (A.M.B., D.W.F., J.E.M.); Drug Safety Research and Development, Pfizer, Inc., New York, New York (S.N.C.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio (S.A.X.)
| | - Sarah N Campion
- Departments of Pharmacology and Toxicology (M.J.C., M.D.M., R.N.H., N.J.C.) and Pediatrics (H.A.K., R.P.E.), University of Arizona, Tucson, Arizona; School of Pharmacy, University of Connecticut, Storrs, Connecticut (A.M.B., D.W.F., J.E.M.); Drug Safety Research and Development, Pfizer, Inc., New York, New York (S.N.C.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio (S.A.X.)
| | - Daniel W Ferreira
- Departments of Pharmacology and Toxicology (M.J.C., M.D.M., R.N.H., N.J.C.) and Pediatrics (H.A.K., R.P.E.), University of Arizona, Tucson, Arizona; School of Pharmacy, University of Connecticut, Storrs, Connecticut (A.M.B., D.W.F., J.E.M.); Drug Safety Research and Development, Pfizer, Inc., New York, New York (S.N.C.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio (S.A.X.)
| | - Stavra A Xanthakos
- Departments of Pharmacology and Toxicology (M.J.C., M.D.M., R.N.H., N.J.C.) and Pediatrics (H.A.K., R.P.E.), University of Arizona, Tucson, Arizona; School of Pharmacy, University of Connecticut, Storrs, Connecticut (A.M.B., D.W.F., J.E.M.); Drug Safety Research and Development, Pfizer, Inc., New York, New York (S.N.C.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio (S.A.X.)
| | - Jose E Manautou
- Departments of Pharmacology and Toxicology (M.J.C., M.D.M., R.N.H., N.J.C.) and Pediatrics (H.A.K., R.P.E.), University of Arizona, Tucson, Arizona; School of Pharmacy, University of Connecticut, Storrs, Connecticut (A.M.B., D.W.F., J.E.M.); Drug Safety Research and Development, Pfizer, Inc., New York, New York (S.N.C.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio (S.A.X.)
| | - H Hesham A-Kader
- Departments of Pharmacology and Toxicology (M.J.C., M.D.M., R.N.H., N.J.C.) and Pediatrics (H.A.K., R.P.E.), University of Arizona, Tucson, Arizona; School of Pharmacy, University of Connecticut, Storrs, Connecticut (A.M.B., D.W.F., J.E.M.); Drug Safety Research and Development, Pfizer, Inc., New York, New York (S.N.C.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio (S.A.X.)
| | - Robert P Erickson
- Departments of Pharmacology and Toxicology (M.J.C., M.D.M., R.N.H., N.J.C.) and Pediatrics (H.A.K., R.P.E.), University of Arizona, Tucson, Arizona; School of Pharmacy, University of Connecticut, Storrs, Connecticut (A.M.B., D.W.F., J.E.M.); Drug Safety Research and Development, Pfizer, Inc., New York, New York (S.N.C.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio (S.A.X.)
| | - Nathan J Cherrington
- Departments of Pharmacology and Toxicology (M.J.C., M.D.M., R.N.H., N.J.C.) and Pediatrics (H.A.K., R.P.E.), University of Arizona, Tucson, Arizona; School of Pharmacy, University of Connecticut, Storrs, Connecticut (A.M.B., D.W.F., J.E.M.); Drug Safety Research and Development, Pfizer, Inc., New York, New York (S.N.C.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio (S.A.X.)
| |
Collapse
|
28
|
Li D, Wang X, Lan X, Li Y, Liu L, Yi J, Li J, Sun Q, Wang Y, Li H, Zhong N, Holmdahl R, Lu S. Down-regulation of miR-144 elicits proinflammatory cytokine production by targeting toll-like receptor 2 in nonalcoholic steatohepatitis of high-fat-diet-induced metabolic syndrome E3 rats. Mol Cell Endocrinol 2015; 402:1-12. [PMID: 25534427 DOI: 10.1016/j.mce.2014.12.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2014] [Revised: 12/06/2014] [Accepted: 12/09/2014] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To analyze regulatory microRNA(s) leading to increased TLR2 expression in livers of high-fat-diet induced metabolic syndrome (HFD-MetS) in rats with non-alcoholic steatohepatitis (NASH). METHODS TLRs, inflammatory cytokines, candidate miRNAs targeting key TLR and its cellular localization were determined in liver. The miR-144 targeting TLR2 and regulating TLR2 signaling were further determined by dual luciferase reporter assay and miR-144 mimics or inhibitor. RESULTS Expression of miR-144 was negatively correlated with TLR2 expression in Kupffer cells. The miR-144 bound to 3'UTR of rat TLR2 mRNA. In addition, compared to control group, TLR2, TNF-α, IFN-γ and activation of NF-κB decreased after miR-144 mimic challenge in NR8383 cells and BMM from E3 rats, which could be compensated by Pam3CSK4; while opposite effects on their expressions were observed after miR-144 inhibitor administration, augmented by Pam3CSK4. CONCLUSION Decreased miR-144 could enhance TNF-α and IFN-γ production by targeting TLR2 in vitro, and might contribute to TLR2 up-regulation and the progression of NASH in HFD-MetS E3 rats. This might offer a novel and potential target for NASH therapy.
Collapse
Affiliation(s)
- Dongmin Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Beijing, China
| | - Xuan Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Beijing, China
| | - Xi Lan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Beijing, China
| | - Yue Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Beijing, China
| | - Li Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Beijing, China
| | - Jing Yi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Beijing, China
| | - Jing Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Beijing, China
| | - Qingzhu Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Beijing, China
| | - Yili Wang
- Research Institute of Cancer, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Hongmin Li
- School of Life Sciences, Northwest University, Xi'an, Shaanxi 710061, China
| | - Nannan Zhong
- Xi'an Health School, Xi'an, Shaanxi 710054, China
| | - Rikard Holmdahl
- Division of Medical Inflammation Research, Department of Biochemistry and Biophysics, Karolinska Institute, SE-17177 Stockholm, Sweden
| | - Shemin Lu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Beijing, China.
| |
Collapse
|
29
|
Canet MJ, Hardwick RN, Lake AD, Dzierlenga AL, Clarke JD, Goedken MJ, Cherrington NJ. Renal xenobiotic transporter expression is altered in multiple experimental models of nonalcoholic steatohepatitis. Drug Metab Dispos 2014; 43:266-72. [PMID: 25488932 DOI: 10.1124/dmd.114.060574] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Nonalcoholic fatty liver disease is the most common chronic liver disease, which can progress to nonalcoholic steatohepatitis (NASH). Previous investigations demonstrated alterations in the expression and activity of hepatic drug transporters in NASH. Moreover, studies using rodent models of cholestasis suggest that compensatory changes in kidney transporter expression occur to facilitate renal excretion during states of hepatic stress; however, little information is currently known regarding extrahepatic regulation of drug transporters in NASH. The purpose of the current study was to investigate the possibility of renal drug transporter regulation in NASH across multiple experimental rodent models. Both rat and mouse NASH models were used in this investigation and include: the methionine and choline-deficient (MCD) diet, atherogenic diet, fa/fa rat, ob/ob and db/db mice. Histologic and pathologic evaluations confirmed that the MCD and atherogenic rats as well as the ob/ob and db/db mice all developed NASH. In contrast, the fa/fa rats did not develop NASH but did develop extensive renal injury compared with the other models. Renal mRNA and protein analyses of xenobiotic transporters suggest that compensatory changes occur in NASH to favor increased xenobiotic secretion. Specifically, both apical efflux and basolateral uptake transporters are induced, whereas apical uptake transporter expression is repressed. These results suggest that NASH may alter the expression and potentially function of renal drug transporters, thereby impacting drug elimination mechanisms in the kidney.
Collapse
Affiliation(s)
- Mark J Canet
- University of Arizona, Department of Pharmacology and Toxicology, Tucson, Arizona (M.J.C., R.N.H., A.D.L., A.L.D., J.D.C., N.J.C.); and Rutgers University, Office of Translational Science, New Brunswick, New Jersey (M.J.G.)
| | - Rhiannon N Hardwick
- University of Arizona, Department of Pharmacology and Toxicology, Tucson, Arizona (M.J.C., R.N.H., A.D.L., A.L.D., J.D.C., N.J.C.); and Rutgers University, Office of Translational Science, New Brunswick, New Jersey (M.J.G.)
| | - April D Lake
- University of Arizona, Department of Pharmacology and Toxicology, Tucson, Arizona (M.J.C., R.N.H., A.D.L., A.L.D., J.D.C., N.J.C.); and Rutgers University, Office of Translational Science, New Brunswick, New Jersey (M.J.G.)
| | - Anika L Dzierlenga
- University of Arizona, Department of Pharmacology and Toxicology, Tucson, Arizona (M.J.C., R.N.H., A.D.L., A.L.D., J.D.C., N.J.C.); and Rutgers University, Office of Translational Science, New Brunswick, New Jersey (M.J.G.)
| | - John D Clarke
- University of Arizona, Department of Pharmacology and Toxicology, Tucson, Arizona (M.J.C., R.N.H., A.D.L., A.L.D., J.D.C., N.J.C.); and Rutgers University, Office of Translational Science, New Brunswick, New Jersey (M.J.G.)
| | - Michael J Goedken
- University of Arizona, Department of Pharmacology and Toxicology, Tucson, Arizona (M.J.C., R.N.H., A.D.L., A.L.D., J.D.C., N.J.C.); and Rutgers University, Office of Translational Science, New Brunswick, New Jersey (M.J.G.)
| | - Nathan J Cherrington
- University of Arizona, Department of Pharmacology and Toxicology, Tucson, Arizona (M.J.C., R.N.H., A.D.L., A.L.D., J.D.C., N.J.C.); and Rutgers University, Office of Translational Science, New Brunswick, New Jersey (M.J.G.)
| |
Collapse
|
30
|
Bao Y, Mo J, Ruan L, Li G. Increased monocytic CD14⁺HLADRlow/- myeloid-derived suppressor cells in obesity. Mol Med Rep 2014; 11:2322-8. [PMID: 25384365 DOI: 10.3892/mmr.2014.2927] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 10/24/2014] [Indexed: 11/05/2022] Open
Abstract
Obesity is associated with numerous immunological disorders. The present study investigated the proportion and phenotype of myeloid‑derived suppressor cells (MDSCs) in the plasma of obese subjects and the association of these cells with the level of liver enzymes. Certain features of the immune response in obese subjects were examined by analyzing the expression of T cell receptor‑ζ (TCRζ) molecules on the surface of T cells. The expression and secretion of S100A9, a possible marker for MDSCs, were detected in the peripheral blood of obese subjects and compared with levels in lean controls. Results showed that the percentage of monocytic MDSCs, with the phenotype CD33+CD11b+CD14+HLADRlow/‑, was significantly increased in obese subjects compared with lean controls. The circulating level of monocytic MDSCs was positively correlated with the levels of liver enzymes in serum. The expression of the TCRζ molecule in the resting T cells was significantly lower in obese individuals than that of lean controls. The expression of S100A9 was detected in the majority of monocytes in peripheral blood mononulear cells, but no difference was identified in the frequency of CD14+S100A9+ cells between the obese and lean groups. However, the plasma level of S100A8/9 was significantly increased in obese compared with lean subjects. These observations suggested that the increased frequency of CD33+CD11b+CD14+HLADRlow/‑ cells may be responsible for the impaired T‑cell function and liver injury observed in obesity.
Collapse
Affiliation(s)
- Yi Bao
- Department of Basic and Translational Research, The Key Laboratory, The Second Hospital of Jiaxing, Jiaxing, Zhejiang 314000, P.R. China
| | - Juanfen Mo
- Department of Basic and Translational Research, The Key Laboratory, The Second Hospital of Jiaxing, Jiaxing, Zhejiang 314000, P.R. China
| | - Lingjuan Ruan
- Department of Basic and Translational Research, The Key Laboratory, The Second Hospital of Jiaxing, Jiaxing, Zhejiang 314000, P.R. China
| | - Guo Li
- Department of Basic and Translational Research, The Key Laboratory, The Second Hospital of Jiaxing, Jiaxing, Zhejiang 314000, P.R. China
| |
Collapse
|
31
|
Nonalcoholic steatohepatitis associated with metabolic syndrome: relationship to insulin resistance and liver histology. J Clin Gastroenterol 2014; 48:883-8. [PMID: 24440936 DOI: 10.1097/mcg.0000000000000065] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Nonalcoholic steatohepatitis (NASH) is a hepatic manifestation of metabolic syndrome. We aimed to assess the relationship of metabolic syndrome-associated NASH and insulin resistance (IR), and to define the correlation of chemicometabolic components with different degree of liver histology in NASH subjects. STUDY Ninety-four subjects with NASH (mean age, 38±14 y; 77% male) were enrolled. IR was calculated using a homeostasis model assessment of insulin resistance (HOMA-IR). Clinical characteristics including IR and accompanying metabolic risk components in NASH subjects were compared with those of 52 diabetics and 21 healthy controls. The relationship between IR and chemicometabolic variables was analyzed according to different clustering of metabolic risk components and the histologic activity. RESULTS NASH subjects had a stronger association with metabolic syndrome than healthy controls. HOMA-IR was significantly higher in NASH subjects than in healthy controls (4.4±2.5 vs. 1.7±0.6; P<0.001) but not than in diabetics. NASH subjects with metabolic syndrome were more likely to have higher HOMA-IR compared with that of NASH subjects without metabolic syndrome (5.0±2.9 vs. 3.6±1.7; P=0.032). HOMA-IR showed a positive correlation with body mass index (r=0.428, P=0.015) and serum fasting blood sugar (r=0.365, P=0.037). Serum aspartate aminotransferase/alanine aminotransferase ratio (P=0.029) and high-density lipoprotein cholesterol level (P=0.034) were significantly affected according to the degree of fibrotic activity in 41 histology-proven NASH subjects. CONCLUSIONS NASH subjects showed increased IR with a significant association of metabolic syndrome. The severity of hepatic fibrosis revealed a strong correlation with serum aspartate aminotransferase/alanine aminotransferase ratio and high-density lipoprotein cholesterol level.
Collapse
|
32
|
Oliveira VB, Ferreira AV, Oliveira MC, Teixeira MM, Brandão MG. Effects of Xylopia aromatica (Lam.) Mart. fruit on metabolic and inflammatory dysfunction induced by high refined carbohydrate-containing-diet in mice. Food Res Int 2014. [DOI: 10.1016/j.foodres.2014.03.066] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
33
|
Ganz M, Csak T, Szabo G. High fat diet feeding results in gender specific steatohepatitis and inflammasome activation. World J Gastroenterol 2014; 20:8525-8534. [PMID: 25024607 PMCID: PMC4093702 DOI: 10.3748/wjg.v20.i26.8525] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 02/28/2013] [Accepted: 06/04/2013] [Indexed: 02/06/2023] Open
Abstract
AIM: To develop an animal model that encompasses the different facets of non-alcoholic steatohepatitis (NASH), which has been a challenge.
METHODS: In this study, we used a high fat diet (HFD) feeding supplemented with fructose and sucrose in the water mimicking the high-fructose corn syrup that is abundant in the diet in the United States. We used C57Bl/6 wild-type mice for short and long-term feedings of 6 and 16 wk respectively, and evaluated the extent of liver damage, steatosis, and inflammasome activation. Our methods included histopathological analysis to assess liver damage and steatosis, which involved H and E and oil-red-o staining; biochemical studies to look at ALT and triglyceride levels; RNA analysis using quantitative polymerase chain reaction; and cytokine analysis, which included the enzyme-linked immunosorbent assay method to look at interleukin (IL)-1β and tumor necrosis factor-α (TNFα) levels. Furthermore, at each length of feeding we also looked at insulin resistance and glucose tolerance using insulin tolerance tests (ITT) and glucose tolerance tests.
RESULTS: There was no insulin resistance, steatosis, or inflammasome activation at 6 wk. In contrast, at 16 wk we found significant insulin resistance demonstrated by impaired glucose and ITT in male, but not female mice. In males, elevated alanine aminotransferase and triglyceride levels, indicated liver damage and steatosis, respectively. Increased liver TNFα and monocyte chemoattractant protein-1 mRNA and protein, correlated with steatohepatitis. The inflammasome components, adaptor molecule, Aim2, and NOD-like receptor 4, increased at the mRNA level, and functional inflammasome activation was indicated by increased caspase-1 activity and IL-1β protein levels in male mice fed a long-term HFD. Male mice on HFD had increased α-smooth muscle actin and pro-collagen-1 mRNA indicating evolving fibrosis. In contrast, female mice displayed only elevated triglyceride levels, steatosis, and no fibrosis.
CONCLUSION: Our data indicate gender differences in NASH. Male mice fed a long-term HFD display steatohepatitis and inflammasome activation, whereas female mice have steatosis without inflammation.
Collapse
|
34
|
Lai YS, Chen WC, Ho CT, Lu KH, Lin SH, Tseng HC, Lin SY, Sheen LY. Garlic essential oil protects against obesity-triggered nonalcoholic fatty liver disease through modulation of lipid metabolism and oxidative stress. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2014; 62:5897-906. [PMID: 24857364 DOI: 10.1021/jf500803c] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
This study investigated the protective properties of garlic essential oil (GEO) and its major organosulfur component (diallyl disulfide, DADS) against the development of nonalcoholic fatty liver disease (NAFLD). C57BL/6J mice were fed a normal or high-fat diet (HFD) with/without GEO (25, 50, and 100 mg/kg) or DADS (10 and 20 mg/kg) for 12 weeks. GEO and DADS dose-dependently exerted antiobesity and antihyperlipidemic effects by reducing HFD-induced body weight gain, adipose tissue weight, and serum biochemical parameters. Administration of 50 and 100 mg/kg GEO and 20 mg/kg DADS significantly decreased the release of pro-inflammatory cytokines in liver, accompanied by elevated antioxidant capacity via inhibition of cytochrome P450 2E1 expression during NAFLD development. The anti-NAFLD effects of GEO and DADS were mediated through down-regulation of sterol regulatory element binding protein-1c, acetyl-CoA carboxylase, fatty acid synthase, and 3-hydroxy-3-methylglutaryl-coenzyme A reductase, as well as stimulation of peroxisome proliferator-activated receptor α and carnitine palmitoyltransferase-1. These results demonstrate that GEO and DADS dose-dependently protected obese mice with long-term HFD-induced NAFLD from lipid accumulation, inflammation, and oxidative damage by ameliorating lipid metabolic disorders and oxidative stress. The dose of 20 mg/kg DADS was equally as effective in preventing NAFLD as 50 mg/kg GEO containing the same amount of DADS, which demonstrates that DADS may be the main bioactive component in GEO.
Collapse
Affiliation(s)
- Yi-Syuan Lai
- Institute of Food Science and Technology, National Taiwan University , Taipei 10617, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW Hepatic encephalopathy causes significant cognitive impairment and morbidity in patients with cirrhosis; however, hepatic encephalopathy is considered a reversible syndrome once recognized clinically. Although hepatic encephalopathy is not a single clinical entity, the pathophysiology resulting in brain dysfunction is not fully understood, although it is believed that ammonia production is an important contributing factor. The purpose of this review is to highlight studies used to test for hepatic encephalopathy and those utilizing specific new treatments. RECENT FINDINGS A 'STROOP' smartphone app has been developed to allow clinicians to test for covert hepatic encephalopathy (CHE). Lactulose therapy was effective for cirrhotic patients as primary prophylaxis to prevent overt hepatic encephalopathy (OHE) episodes. In patients without prior OHE, probiotics can be useful in preventing OHE. Lactulose, probiotics, L-ornithine-L-aspartate, and potassium-iron-phosphate-citrate have been studied in the treatment of CHE. Rifaximin was found to be safe and well tolerated in long-term maintenance of remission from OHE; however, compared to lactulose therapy in CHE, it is not cost-effective. SUMMARY Refinement in clinical management strategies for patients with cirrhosis and hepatic encephalopathy appears to continue to contribute to improved patient outcomes.
Collapse
|
36
|
Mobley CB, Toedebusch RG, Lockwood CM, Heese AJ, Zhu C, Krieger AE, Cruthirds CL, Hofheins JC, Company JM, Wiedmeyer CE, Kim DY, Booth FW, Roberts MD. Herbal adaptogens combined with protein fractions from bovine colostrum and hen egg yolk reduce liver TNF-α expression and protein carbonylation in Western diet feeding in rats. Nutr Metab (Lond) 2014; 11:19. [PMID: 24822076 PMCID: PMC4017828 DOI: 10.1186/1743-7075-11-19] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Accepted: 04/16/2014] [Indexed: 12/13/2022] Open
Abstract
Background We examined if a purported anti-inflammatory supplement (AF) abrogated Western-diet (WD)-induced liver pathology in rats. AF contained: 1) protein concentrates from bovine colostrum and avian egg yolk; 2) herbal adaptogens and antioxidants; and 3) acetyl-L-carnitine. Methods Nine month-old male Brown Norway rats were allowed ad libitum access to WD for 41–43 days and randomly assigned to WD + AF feeding twice daily for the last 31–33 days (n = 8), or WD and water-placebo feeding twice daily for the last 31–33 days (n = 8). Rats fed a low-fat/low-sucrose diet (CTL, n = 6) for 41–43 days and administered a water-placebo twice daily for the last 31–33 days were also studied. Twenty-four hours following the last gavage-feed, liver samples were analyzed for: a) select mRNAs (via RT-PCR) as well as genome-wide mRNA expression patterns (via RNA-seq); b) lipid deposition; and, c) protein carbonyl and total antioxidant capacity (TAC). Serum was also examined for TAC, 8-isoprostane and clinical chemistry markers. Results WD + AF rats experienced a reduction in liver Tnf-α mRNA (-2.8-fold, p < 0.01). Serum and liver TAC was lower in WD + AF versus WD and CTL rats (p < 0.05), likely due to exogenous antioxidant ingredients provided through AF as evidenced by a tendency for mitochondrial SOD2 mRNA to increase in WD + AF versus CTL rats (p = 0.07). Liver fat deposition nor liver protein carbonyl content differed between WD + AF versus WD rats, although liver protein carbonyls tended to be lower in WD + AF versus CTL rats (p = 0.08). RNA-seq revealed that 19 liver mRNAs differed between WD + AF versus WD when both groups were compared with CTL rats (+/- 1.5-fold, p < 0.01). Bioinformatics suggest that AF prevented WD-induced alterations in select genes related to the transport and metabolism of carbohydrates in favor of select genes related to lipid transport and metabolism. Finally, serum clinical safety markers and liver pathology (via lesion counting) suggests that chronic consumption of AF was well tolerated. Conclusions AF supplementation elicits select metabolic, anti-inflammatory, and anti-oxidant properties which was in spite of WD feeding and persisted up to 24 hours after receiving a final dose.
Collapse
Affiliation(s)
| | - Ryan G Toedebusch
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, USA
| | | | - Alexander J Heese
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, USA
| | - Conan Zhu
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, USA
| | - Anna E Krieger
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, USA
| | - Clayton L Cruthirds
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, USA
| | - John C Hofheins
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, USA
| | - Joseph M Company
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, USA
| | - Charles E Wiedmeyer
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
| | - Dae Y Kim
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
| | - Frank W Booth
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, USA ; Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA ; Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA ; Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Michael D Roberts
- School of Kinesiology, Auburn University, Auburn, AL, USA ; Department of Biomedical Sciences, University of Missouri, Columbia, MO, USA
| |
Collapse
|
37
|
Boyraz M, Cekmez F, Karaoglu A, Cinaz P, Durak M, Bideci A. Serum adiponectin, leptin, resistin and RBP4 levels in obese and metabolic syndrome children with nonalcoholic fatty liver disease. Biomark Med 2014; 7:737-45. [PMID: 24044566 DOI: 10.2217/bmm.13.13] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
AIM To investigate the relationship of adiponectin, leptin, resistin and RBP4 levels in obese and metabolic syndrome children with nonalcoholic fatty liver disease (NAFLD). PATIENTS & METHODS Group I consisted of 63 obese children with liver steatosis, group II consisted of 12 obese children with elevated serum ALT activity from group I, and group III included 85 obese children without liver steatosis. RESULTS Leptin levels were higher in the NAFLD children than in the control group. Serum RBP4 levels in obese children with NAFLD were higher than those in obese children without NAFLD and controls. Adiponectin and resistin levels were negatively correlated and RBP4 levels positively correlated with ALT activity and ultrasonographic grading. CONCLUSION These data suggest that adiponectin, resistin and RBP4 may have a role in the pathogenesis of NAFLD in obese children. Adiponectin, leptin, resistin and RBP4 may be suitable markers for predicting metabolic syndrome and NAFLD.
Collapse
Affiliation(s)
- Mehmet Boyraz
- Department of Pediatrics, Division of Endocrinology, Şişli Etfal Education & Training Hospital, Istanbul, Turkey
| | | | | | | | | | | |
Collapse
|
38
|
Kim H, Lee K, Lee KW, Yi NJ, Lee HW, Hong G, Choi Y, You T, Suh SW, Jang JJ, Suh KS. Histologically proven non-alcoholic fatty liver disease and clinically related factors in recipients after liver transplantation. Clin Transplant 2014; 28:521-9. [PMID: 24579874 DOI: 10.1111/ctr.12343] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/13/2014] [Indexed: 12/27/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) affects a substantial proportion of the world population, and its prevalence has been increasing. The study was aimed at evaluating the prevalence and peri-transplant risk factors for post-liver transplantation (LT) NAFLD. A retrospective review was performed for adult recipients who underwent late protocol biopsy (>1 yr after LT) between August 2010 and December 2012. Hepatic steatosis was reviewed and graded by hepatopathologists, and the peri-transplant factors were analyzed for relationships to histologically proven NAFLD. Total 166 biopsies had been performed in 156 recipients. NAFLD was present in 27.1% at a mean period of 35.4 months between LT and biopsy, moderate and severe steatosis (≥33%) consisted of 28.9%. In multivariate analysis, pre-LT alcoholic cirrhosis (odds ratio [OR] 8.031, p = 0.003), obesity at biopsy (OR 3.873, p = 0.001), and preexisting donor graft steatosis (OR 3.147, p = 0.022) were significant risk factors for post-LT NAFLD. In conclusion, NAFLD represented a considerable portion of recipients, but this prevalence was not higher than those for general population. Three risk factors were significantly related to post-LT NAFLD, and recipients with those factors should be monitored for NAFLD. Furthermore, possible progression to non-alcoholic steatohepatitis (NASH) or fibrosis and metabolic syndrome should be considered in future studies.
Collapse
Affiliation(s)
- Hyeyoung Kim
- Department of Surgery, College of Medicine, Seoul National University, Seoul, Republic of Korea; Department of Surgery, Seoul Metropolitan Government, Boramae Medical Center, Seoul National University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Voma C, Barfell A, Croniger C, Romani A. Reduced cellular Mg²⁺ content enhances hexose 6-phosphate dehydrogenase activity and expression in HepG2 and HL-60 cells. Arch Biochem Biophys 2014; 548:11-9. [PMID: 24631573 DOI: 10.1016/j.abb.2014.02.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 02/25/2014] [Accepted: 02/26/2014] [Indexed: 11/16/2022]
Abstract
We have reported that Mg(2+) dynamically regulates glucose 6-phosphate entry into the endoplasmic reticulum and its hydrolysis by the glucose 6-phosphatase in liver cells. In the present study, we report that by modulating glucose 6-phosphate entry into the endoplasmic reticulum of HepG2 cells, Mg(2+) also regulates the oxidation of this substrate via hexose 6-phosphate dehydrogenase (H6PD). This regulatory effect is dynamic as glucose 6-phosphate entry and oxidation can be rapidly down-regulated by the addition of exogenous Mg(2+). In addition, HepG2 cells growing in low Mg(2+) show a marked increase in hexose 6-phosphate dehydrogenase mRNA and protein expression. Metabolically, these effects on hexose 6-phosphate dehydrogenase are important as this enzyme increases intra-reticular NADPH production, which favors fatty acid and cholesterol synthesis. Similar effects of Mg(2+) were observed in HL-60 cells. These and previously published results suggest that in an hepatocyte culture model changes in cytoplasmic Mg(2+) content regulates glucose 6-phosphate utilization via glucose 6 phosphatase and hexose-6 phosphate dehydrogenase in alternative to glycolysis and glycogen synthesis. This alternative regulation might be of relevance in the transition from fed to fasted state.
Collapse
Affiliation(s)
- Chesinta Voma
- Department of Physiology and Biophysics, Case Western Reserve University, USA; Department of Clinical Chemistry, Cleveland State University, USA
| | - Andrew Barfell
- Department of Physiology and Biophysics, Case Western Reserve University, USA
| | | | - Andrea Romani
- Department of Physiology and Biophysics, Case Western Reserve University, USA.
| |
Collapse
|
40
|
Abstract
Non-alcoholic fatty liver disease affects nearly 30% of Americans. A histopathological spectrum exists from simple steatosis to NASH which may progress to cirrhosis and HCC. NASH is currently the third most common indication for liver transplant with increasing incidence. Steatosis can be considered the hepatic manifestation of the metabolic syndrome as insulin resistance is a major risk factor for its development. While liver biopsy is the gold standard for diagnosis, non-invasive methods are currently being developed to appropriately determine who needs histologic evaluation. Management focuses on mitigation of risk factors, since targeted therapies to halt progression of fibrosis have not been validated. Simple steatosis does not affect overall survival, but NASH conveys increased mortality. Because of this, non-invasive strategies to diagnose patients and management algorithms are needed. This review supports the definitions of simple steatosis and NASH as two distinct entities based on pathophysiology, diagnosis, management, and prognosis.
Collapse
|
41
|
Said A. Non-alcoholic fatty liver disease and liver transplantation: Outcomes and advances. World J Gastroenterol 2013; 19:9146-9155. [PMID: 24409043 PMCID: PMC3882389 DOI: 10.3748/wjg.v19.i48.9146] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 10/28/2013] [Accepted: 11/03/2013] [Indexed: 02/07/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the most prevalent causes of chronic liver disease worldwide. In the last decade it has become the third most common indication for liver transplantation in the United States. Increasing prevalence of NAFLD in the general population also poses a risk to organ donation, as allograft steatosis can be associated with non-function of the graft. Post-transplant survival is comparable between NAFLD and non-NAFLD causes of liver disease, although long term outcomes beyond 10 year are lacking. NAFLD can recur in the allograft frequently although thus far post transplant survival has not been impacted. De novo NAFLD can also occur in the allograft of patients transplanted for non-NAFLD liver disease. Predictors for NAFLD post-transplant recurrence include obesity, hyperlipidemia and diabetes as well as steroid dose after liver transplantation. A polymorphism in PNPLA3 that mediates triglyceride hydrolysis and is linked to pre-transplant risk of obesity and NAFLD has also been linked to post transplant NAFLD risk. Although immunosuppression side effects potentiate obesity and the metabolic syndrome, studies of immunosuppression modulation and trials of specific immunosuppression regimens post-transplant are lacking in this patient population. Based on pre-transplant data, sustained weight loss through diet and exercise is the most effective therapy for NAFLD. Other agents occasionally utilized in NAFLD prior to transplantation include vitamin E and insulin-sensitizing agents. Studies of these therapies are lacking in the post-transplant population. A multimodality and multidisciplinary approach to treatment should be utilized in management of post-transplant NAFLD.
Collapse
|
42
|
Savini I, Catani MV, Evangelista D, Gasperi V, Avigliano L. Obesity-associated oxidative stress: strategies finalized to improve redox state. Int J Mol Sci 2013; 14:10497-538. [PMID: 23698776 PMCID: PMC3676851 DOI: 10.3390/ijms140510497] [Citation(s) in RCA: 327] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2013] [Revised: 04/18/2013] [Accepted: 05/06/2013] [Indexed: 12/14/2022] Open
Abstract
Obesity represents a major risk factor for a plethora of severe diseases, including diabetes, cardiovascular disease, non-alcoholic fatty liver disease, and cancer. It is often accompanied by an increased risk of mortality and, in the case of non-fatal health problems, the quality of life is impaired because of associated conditions, including sleep apnea, respiratory problems, osteoarthritis, and infertility. Recent evidence suggests that oxidative stress may be the mechanistic link between obesity and related complications. In obese patients, antioxidant defenses are lower than normal weight counterparts and their levels inversely correlate with central adiposity; obesity is also characterized by enhanced levels of reactive oxygen or nitrogen species. Inadequacy of antioxidant defenses probably relies on different factors: obese individuals may have a lower intake of antioxidant- and phytochemical-rich foods, such as fruits, vegetables, and legumes; otherwise, consumption of antioxidant nutrients is normal, but obese individuals may have an increased utilization of these molecules, likewise to that reported in diabetic patients and smokers. Also inadequate physical activity may account for a decreased antioxidant state. In this review, we describe current concepts in the meaning of obesity as a state of chronic oxidative stress and the potential interventions to improve redox balance.
Collapse
Affiliation(s)
- Isabella Savini
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy.
| | | | | | | | | |
Collapse
|
43
|
Abstract
PURPOSE OF REVIEW Chronic liver disease causes significant morbidity and mortality because of any number of complications including hepatic encephalopathy, ascites, hepatorenal syndrome (HRS), and esophageal variceal hemorrhage (EVH). RECENT FINDINGS Predictors of response to lactulose, probiotics, and L-ornithine-L-aspartate therapy in minimal hepatic encephalopathy (MHE) have been reported. Although rifaximin was slightly more effective than lactulose in the maintenance of remission and decreased re-admission in patients with MHE, it was not as cost-effective as lactulose. Beta-blockade has been associated with paracentesis-induced circulatory dysfunction. Those who respond to nonselective beta-blockers have a predictable overall lower probability of developing ascites and HRS. Noradrenaline was as effective as terlipressin for the treatment of type 1 HRS and was less costly. Hemorrhagic ascites, defined as an ascitic fluid red blood cell (RBC) count of at least 10 000/μl, appeared to be a marker for poor outcome in patients with cirrhosis. In patients with acute EVH, band ligation, pharmacologic vasoconstrictors, and antibiotics are effective; notably, intravenous proton pump inhibitor therapy in lieu of vasoconstrictors achieved similar hemostatic effects with fewer side-effects. SUMMARY Refinement in the clinical management strategies for patients with cirrhosis and its complications appear to continue to contribute to improved patient outcomes.
Collapse
|
44
|
|