751
|
Gao T, Liu T, Ko CJ, Zhang L, Joo D, Xie X, Zhu L, Li Y, Cheng X, Sun SC. Myeloid cell TBK1 restricts inflammatory responses. Proc Natl Acad Sci U S A 2022; 119:e2107742119. [PMID: 35074921 PMCID: PMC8794809 DOI: 10.1073/pnas.2107742119] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 11/23/2021] [Indexed: 12/21/2022] Open
Abstract
Proinflammatory cytokine production by innate immune cells plays a crucial role in inflammatory diseases, but the molecular mechanisms controlling the inflammatory responses are poorly understood. Here, we show that TANK-binding kinase 1 (TBK1) serves as a vital regulator of proinflammatory macrophage function and protects against tissue inflammation. Myeloid cell-conditional Tbk1 knockout (MKO) mice spontaneously developed adipose hypertrophy and metabolic disorders at old ages, associated with increased adipose tissue M1 macrophage infiltration and proinflammatory cytokine expression. When fed with a high-fat diet, the Tbk1-MKO mice also displayed exacerbated hepatic inflammation and insulin resistance, developing symptoms of nonalcoholic steatohepatitis. Furthermore, myeloid cell-specific TBK1 ablation exacerbates inflammation in experimental colitis. Mechanistically, TBK1 functions in macrophages to suppress the NF-κB and MAP kinase signaling pathways and thus attenuate induction of proinflammatory cytokines, particularly IL-1β. Ablation of IL-1 receptor 1 (IL-1R1) eliminates the inflammatory symptoms of Tbk1-MKO mice. These results establish TBK1 as a pivotal anti-inflammatory mediator that restricts inflammation in different disease models.
Collapse
Affiliation(s)
- Tianxiao Gao
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Ting Liu
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Chun-Jung Ko
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030;
- Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei, Taiwan 100233
| | - Lingyun Zhang
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Donghyun Joo
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Xiaoping Xie
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Lele Zhu
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Yanchuan Li
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Xuhong Cheng
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Shao-Cong Sun
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030;
- MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, TX 77030
| |
Collapse
|
752
|
Heo S, Kim DW, Choi SH, Kim SW, Jang JK. Diagnostic performance of liver fibrosis assessment by quantification of liver surface nodularity on computed tomography and magnetic resonance imaging: systematic review and meta-analysis. Eur Radiol 2022; 32:3377-3387. [DOI: 10.1007/s00330-021-08436-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/23/2021] [Accepted: 10/24/2021] [Indexed: 12/14/2022]
|
753
|
Hepatocellular BChE as a therapeutic target to ameliorate hypercholesterolemia through PRMT5 selective degradation to restore LDL receptor transcription. Life Sci 2022; 293:120336. [PMID: 35065166 DOI: 10.1016/j.lfs.2022.120336] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 01/03/2022] [Accepted: 01/14/2022] [Indexed: 11/22/2022]
Abstract
AIMS Individuals with nonalcoholic hepatosteatosis (NAFLD) have a worse atherogenic lipoprotein profile and are susceptible to cardiovascular diseases. The MEK-ERK signaling cascades are central regulators of the levels of LDL receptor (LDLR), a major determinant of circulating cholesterol. It is elusive how hepatic steatosis contributes to dyslipidemia, especially hypercholesterolemia. MAIN METHODS The effects of BChE on signaling pathways were determined by immunoblotting in a BChE knockout hepatocyte cell line. DiI-LDL probe was used to explore the effect of BChE expression on LDL internalization. Co-immunoprecipitation and LC-MS were used to explore the interacting proteins with BChE. Finally, a hepatocyte-restricted BChE silencing mouse model was established by AAV8-Tbg-shRNA, and the hypercholesterolemia was induced by 65% kcal% high-fat, high-sucrose diet feeding. MAIN FINDINGS Here we demonstrate that butyrylcholinesterase (BChE) governs the LDL receptor levels and LDL uptake capacity through the MEK-ERK signaling cascades to promote Ldlr transcription. BChE interacts and co-localizes with PRMT5, a protein methylation modifier controlling the ERK signaling. PRMT5 regulates LDLR-dependent LDL uptake and is a substrate of chaperone-mediated autophagy (CMA). BChE deficiency induces the PRTM5 degradation dependent on CMA activity, possibly through facilitating the HSC70 (Heat shock cognate 71 kDa) recognition of PRMT5. Remarkably, in vivo hepatocyte-restricted BChE silencing reduces plasma cholesterol levels substantially. In contrast, the BChE knockout mice are predisposed to hypercholesterolemia. SIGNIFICANCE Taken together, these findings outline a regulatory role for the BChE-PRMT5-ERK-LDLR axis in hepatocyte cholesterol metabolism, and suggest that targeting liver BChE is an effective therapeutic strategy to treat hypercholesterolemia.
Collapse
|
754
|
Yu Y, Yu L, Cheng N, Liu X, Fang C, Liu S, Zhu L. Exercise Alleviates the Apolipoprotein A5-Toll-Like Receptor 4 Axis Impairment in Mice With High-Fat Diet-Induced Non-alcoholic Steatohepatitis. Front Physiol 2022; 12:783341. [PMID: 35035359 PMCID: PMC8758581 DOI: 10.3389/fphys.2021.783341] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 11/29/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Apolipoprotein A5 (ApoA5), an important modulator of plasma and hepatic triglyceride metabolism, has been found to be downregulated by metformin to improve non-alcoholic fatty liver disease. Meanwhile, exercise has been recommended as a therapeutic strategy for non-alcoholic steatohepatitis (NASH). However, no study has yet determined whether exercise affects hepatic ApoA5 expression or the inhibition of ApoA5 to toll-like receptor 4 (TLR4). We herein examined the effects of exercise on hepatic ApoA5 expression and the relevance of ApoA5 and TLR4-mediated pathway in mice with high-fat diet (HFD)-induced NASH. Methods: Male C57BL/6J mice were built NASH model with high-fat diet for 12 weeks, and following mice were subjected to exercise for 12 weeks on a treadmill. Microscopy and enzyme-linked immunosorbent assay were used to measure histological analysis of liver and hepatic lipids, respectively. Quantitative real-time PCR and western blot were used to determined mRNA and protein levels of ApoA5 and TLR4-mediated nuclear factor kappa B (NF-κB) pathway components, respectively. ApoA5 overexpression plasmids transfected into mice to investigate the relevance of ApoA5 and TLR4. Results: 12 weeks of exercise remarkably alleviated HFD-induced hepatic lipid accumulation, inflammation, and fibrosis, as well as reduced serum lipopolysaccharide (LPS), hepatic TLR4, myeloid differentiation factor 88 (MyD88), and NF-κBp65 expression. Importantly, exercise did not reduce ApoA5 expression but instead enhanced its ability to suppress TLR4-mediated NF-κB pathway components by decreasing circulating LPS in our experiments involving transfection of ApoA5 overexpression plasmids and LPS interventions. Conclusion: The results demonstrated that exercise improved HFD-induced NASH by triggering the inhibitory effects of ApoA5 on the TLR4-mediated NF-κB pathway.
Collapse
Affiliation(s)
- Yang Yu
- Department of Sports and Health, Guangzhou Sport University, Guangzhou, China
| | - Lina Yu
- Department of Preventive Dentistry, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Nuo Cheng
- Graduate School of Guangzhou Sport University, Guangzhou, China
| | - Xiaoguang Liu
- Department of Sports and Health, Guangzhou Sport University, Guangzhou, China
| | - Chunlu Fang
- Center for Scientific Research and Institute of Exercise and Health, Guangzhou Sport University, Guangzhou, China
| | - Shujing Liu
- Center for Scientific Research and Institute of Exercise and Health, Guangzhou Sport University, Guangzhou, China
| | - Lin Zhu
- Department of Sports and Health, Guangzhou Sport University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Guangzhou Sport University, Guangzhou, China
| |
Collapse
|
755
|
Um YJ, Chang Y, Jung HS, Cho IY, Shin JH, Shin H, Wild SH, Byrne CD, Ryu S. Decrease in Sleep Duration and Poor Sleep Quality over Time Is Associated with an Increased Risk of Incident Non-Alcoholic Fatty Liver Disease. J Pers Med 2022; 12:jpm12010092. [PMID: 35055407 PMCID: PMC8777783 DOI: 10.3390/jpm12010092] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/29/2021] [Accepted: 01/06/2022] [Indexed: 02/04/2023] Open
Abstract
The impact of changes in sleep duration and sleep quality over time on the risk of non-alcoholic fatty liver disease (NAFLD) is not known. We investigated whether changes in sleep duration and in sleep quality between baseline and follow-up are associated with the risk of developing incident NAFLD. The cohort study included 86,530 Korean adults without NAFLD and with a low fibrosis score at baseline. The median follow-up was 3.6 years. Sleep duration and quality were assessed using the Pittsburgh Sleep Quality Index. Hepatic steatosis (HS) and liver fibrosis were assessed using ultrasonography and the fibrosis-4 index (FIB-4). Cox proportional hazard models were used to determine hazard ratios (HRs) and 95% confidence intervals (Cis). A total of 12,127 subjects with incident HS and 559 with incident HS plus intermediate/high FIB-4 was identified. Comparing the decrease in sleep duration of >1 h, with stable sleep duration, the multivariate-adjusted HR (95% CIs) for incident HS was 1.24 (1.15–1.35). The corresponding HRs for incident HS plus intermediate/high FIB-4 was 1.58 (1.10–2.29). Comparing persistently poor sleep quality with persistently good sleep quality, the multivariate-adjusted HR for incident HS was 1.13 (95% CI, 1.05–1.20). A decrease in sleep duration or poor sleep quality over time was associated with an increased risk of incident NAFLD, underscoring an important potential role for good sleep in preventing NAFLD risk.
Collapse
Affiliation(s)
- Yoo Jin Um
- Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 04514, Korea; (Y.J.U.); (H.-S.J.); (I.Y.C.); (J.H.S.); (H.S.)
| | - Yoosoo Chang
- Center for Cohort Studies, Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 04514, Korea
- Department of Occupational and Environmental Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 03181, Korea
- Department of Clinical Research Design & Evaluation, SAIHST, Sungkyunkwan University, Seoul 06355, Korea
- Correspondence: (Y.C.); (S.R.)
| | - Hyun-Suk Jung
- Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 04514, Korea; (Y.J.U.); (H.-S.J.); (I.Y.C.); (J.H.S.); (H.S.)
- Center for Cohort Studies, Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 04514, Korea
| | - In Young Cho
- Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 04514, Korea; (Y.J.U.); (H.-S.J.); (I.Y.C.); (J.H.S.); (H.S.)
- Department of Family Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 03181, Korea
| | - Jun Ho Shin
- Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 04514, Korea; (Y.J.U.); (H.-S.J.); (I.Y.C.); (J.H.S.); (H.S.)
- Department of Surgery, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 03181, Korea
| | - Hocheol Shin
- Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 04514, Korea; (Y.J.U.); (H.-S.J.); (I.Y.C.); (J.H.S.); (H.S.)
- Department of Family Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 03181, Korea
| | - Sarah H. Wild
- Usher Institute, University of Edinburgh, Edinburgh EH8 9AG, UK;
| | - Christopher D Byrne
- Nutrition and Metabolism, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK;
- National Institute for Health Research Southampton Biomedical Research Centre, University Hospital Southampton, Southampton SO16 6YD, UK
| | - Seungho Ryu
- Center for Cohort Studies, Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 04514, Korea
- Department of Occupational and Environmental Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 03181, Korea
- Department of Clinical Research Design & Evaluation, SAIHST, Sungkyunkwan University, Seoul 06355, Korea
- Correspondence: (Y.C.); (S.R.)
| |
Collapse
|
756
|
Laurindo LF, Barbalho SM, Guiguer EL, da Silva Soares de Souza M, de Souza GA, Fidalgo TM, Araújo AC, de Souza Gonzaga HF, de Bortoli Teixeira D, de Oliveira Silva Ullmann T, Sloan KP, Sloan LA. GLP-1a: Going beyond Traditional Use. Int J Mol Sci 2022; 23:739. [PMID: 35054924 PMCID: PMC8775408 DOI: 10.3390/ijms23020739] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 12/28/2021] [Accepted: 12/30/2021] [Indexed: 02/07/2023] Open
Abstract
Glucagon-like peptide-1 (GLP-1) is a human incretin hormone derived from the proglucagon molecule. GLP-1 receptor agonists are frequently used to treat type 2 diabetes mellitus and obesity. However, the hormone affects the liver, pancreas, brain, fat cells, heart, and gastrointestinal tract. The objective of this study was to perform a systematic review on the use of GLP-1 other than in treating diabetes. PubMed, Cochrane, and Embase were searched, and the PRISMA guidelines were followed. Nineteen clinical studies were selected. The results showed that GLP-1 agonists can benefit defined off-medication motor scores in Parkinson's Disease and improve emotional well-being. In Alzheimer's disease, GLP-1 analogs can improve the brain's glucose metabolism by improving glucose transport across the blood-brain barrier. In depression, the analogs can improve quality of life and depression scales. GLP-1 analogs can also have a role in treating chemical dependency, inhibiting dopaminergic release in the brain's reward centers, decreasing withdrawal effects and relapses. These medications can also improve lipotoxicity by reducing visceral adiposity and decreasing liver fat deposition, reducing insulin resistance and the development of non-alcoholic fatty liver diseases. The adverse effects are primarily gastrointestinal. Therefore, GLP-1 analogs can benefit other conditions besides traditional diabetes and obesity uses.
Collapse
Affiliation(s)
- Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília, Avenida Higino Muzzi Filho, Marília 17525-902, SP, Brazil; (L.F.L.); (E.L.G.); (M.d.S.S.d.S.); (G.A.d.S.); (A.C.A.); (H.F.d.S.G.); (T.d.O.S.U.)
| | - Sandra Maria Barbalho
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília, Avenida Higino Muzzi Filho, Marília 17525-902, SP, Brazil; (L.F.L.); (E.L.G.); (M.d.S.S.d.S.); (G.A.d.S.); (A.C.A.); (H.F.d.S.G.); (T.d.O.S.U.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília, Marília 17525-902, SP, Brazil
- Department of Biochemistry and Nutrition, School of Food and Technology of Marilia (FATEC), Marília 17500-000, SP, Brazil
| | - Elen Landgraf Guiguer
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília, Avenida Higino Muzzi Filho, Marília 17525-902, SP, Brazil; (L.F.L.); (E.L.G.); (M.d.S.S.d.S.); (G.A.d.S.); (A.C.A.); (H.F.d.S.G.); (T.d.O.S.U.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília, Marília 17525-902, SP, Brazil
- Department of Biochemistry and Nutrition, School of Food and Technology of Marilia (FATEC), Marília 17500-000, SP, Brazil
| | - Maricelma da Silva Soares de Souza
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília, Avenida Higino Muzzi Filho, Marília 17525-902, SP, Brazil; (L.F.L.); (E.L.G.); (M.d.S.S.d.S.); (G.A.d.S.); (A.C.A.); (H.F.d.S.G.); (T.d.O.S.U.)
| | - Gabriela Achete de Souza
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília, Avenida Higino Muzzi Filho, Marília 17525-902, SP, Brazil; (L.F.L.); (E.L.G.); (M.d.S.S.d.S.); (G.A.d.S.); (A.C.A.); (H.F.d.S.G.); (T.d.O.S.U.)
| | - Thiago Marques Fidalgo
- Department of Psychiatry, Federal University of São Paulo, R. Sena Madureira 04021-001, SP, Brazil;
| | - Adriano Cressoni Araújo
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília, Avenida Higino Muzzi Filho, Marília 17525-902, SP, Brazil; (L.F.L.); (E.L.G.); (M.d.S.S.d.S.); (G.A.d.S.); (A.C.A.); (H.F.d.S.G.); (T.d.O.S.U.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília, Marília 17525-902, SP, Brazil
| | - Heron F. de Souza Gonzaga
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília, Avenida Higino Muzzi Filho, Marília 17525-902, SP, Brazil; (L.F.L.); (E.L.G.); (M.d.S.S.d.S.); (G.A.d.S.); (A.C.A.); (H.F.d.S.G.); (T.d.O.S.U.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília, Marília 17525-902, SP, Brazil
| | - Daniel de Bortoli Teixeira
- Postgraduate Program in Animal Health, Production and Environment, University of Marilia, Marília 17525-902, SP, Brazil;
| | - Thais de Oliveira Silva Ullmann
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília, Avenida Higino Muzzi Filho, Marília 17525-902, SP, Brazil; (L.F.L.); (E.L.G.); (M.d.S.S.d.S.); (G.A.d.S.); (A.C.A.); (H.F.d.S.G.); (T.d.O.S.U.)
| | - Katia Portero Sloan
- Texas Institute for Kidney and Endocrine Disorders, Lufkin, TX 75904, USA; (K.P.S.); (L.A.S.)
| | - Lance Alan Sloan
- Texas Institute for Kidney and Endocrine Disorders, Lufkin, TX 75904, USA; (K.P.S.); (L.A.S.)
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
757
|
Non-Alcoholic Steatohepatitis (NASH) and Organokines: What Is Now and What Will Be in the Future. Int J Mol Sci 2022; 23:ijms23010498. [PMID: 35008925 PMCID: PMC8745668 DOI: 10.3390/ijms23010498] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 12/27/2021] [Accepted: 12/28/2021] [Indexed: 02/05/2023] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is characterized by steatosis, lobular inflammation, and enlargement of the diameter of hepatocytes (ballooning hepatocytes), with or without fibrosis. It affects 20% of patients with non-alcoholic fatty liver disease (NAFLD). Due to liver dysfunction and the numerous metabolic changes that commonly accompany the condition (obesity, insulin resistance, type 2 diabetes, and metabolic syndrome), the secretion of organokines is modified, which may contribute to the pathogenesis or progression of the disease. In this sense, this study aimed to perform a review of the role of organokines in NASH. Thus, by combining descriptors such as NASH, organokines, oxidative stress, inflammation, insulin resistance, and dyslipidemia, a search was carried out in the EMBASE, MEDLINE-PubMed, and Cochrane databases of articles published in the last ten years. Insulin resistance, inflammation and mitochondrial dysfunction, fructose, and intestinal microbiota were factors identified as participating in the genesis and progression of NASH. Changes in the pattern of organokines secretion (adipokines, myokines, hepatokines, and osteokines) directly or indirectly contribute to aggravating the condition or compromise homeostasis. Thus, further studies involving skeletal muscle, adipose, bone, and liver tissue as endocrine organs are essential to better understand the modulation of organokines involved in the pathogenesis of NASH to advance in the treatment of this disease.
Collapse
|
758
|
Wang Q, Wang Z, Pang B, Zheng H, Cao Z, Feng C, Ma W, Wei J. Probiotics for the improvement of metabolic profiles in patients with metabolic-associated fatty liver disease: A systematic review and meta-analysis of randomized controlled trials. Front Endocrinol (Lausanne) 2022; 13:1014670. [PMID: 36407321 PMCID: PMC9670148 DOI: 10.3389/fendo.2022.1014670] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVE This meta-analysis of randomized controlled trials (RCTs) was conducted to assess the efficacy of probiotics in the treatment of metabolic-associated fatty liver disease (MAFLD) mainly in terms of liver function, glucose and lipid metabolism, and inflammation. METHODS RCTs were searched on PubMed, Web of Science, Embase, and the Cochrane Library until June 2022. A meta-analysis was performed on the therapeutic efficacy of probiotics on liver function, glucose and lipid metabolism, and inflammatory biomarkers by using RevMan 5.4 software. RESULTS A total of 772 patients from 15 studies were included in the analysis. The methodological quality varied across studies. We found that adding probiotic therapies could reduce the levels of alanine aminotransferase [mean difference (MD): -11.76 (-16.06, -7.46), p < 0.00001], aspartate aminotransferase (MD: -9.08 (-13.60, -4.56), p < 0.0001], γ-glutamyltransferase [MD: -5.67 (-6.80, -4.54), p < 0.00001] and homeostasis model assessment-insulin resistance [MD: -0.62 (-1.08, -0.15), p = 0.01], in patients with MAFLD compared with those in control individuals. However, there was no statistically significant improvement in the levels of total cholesterol, triglycerides, low-density lipoprotein cholesterol, C-reactive protein and tumor necrosis factor α among patients with MAFLD. Subgroup analyses showed that other key factors, such as age, participants' baseline body mass index, and the duration of intervention, may influence probiotic therapy outcomes. CONCLUSION There is promising evidence that probiotic supplementation can reduce liver enzyme levels and regulate glycometabolism in patients with MAFLD. Further rigorous and long-term trials exploring these novel therapeutic perspectives are warranted to confirm these results.
Collapse
Affiliation(s)
- Qiuhong Wang
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Junping Wei, ; Qiuhong Wang,
| | - Ze Wang
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Boxian Pang
- Graduate school, Beijing University of Chinese Medicine, Beijing, China
| | - Huijuan Zheng
- Renal Research Institution of Beijing University of Chinese Medicine, and Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Zhengmin Cao
- Infections Disease Section, Guang’anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing, China
| | - Chunpeng Feng
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wenxin Ma
- Centre for Evidence-based Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Junping Wei
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Junping Wei, ; Qiuhong Wang,
| |
Collapse
|
759
|
Zhu X, Xia M, Gao X. Update on genetics and epigenetics in metabolic associated fatty liver disease. Ther Adv Endocrinol Metab 2022; 13:20420188221132138. [PMID: 36325500 PMCID: PMC9619279 DOI: 10.1177/20420188221132138] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 09/25/2022] [Indexed: 11/06/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is becoming the most frequent chronic liver disease worldwide. Metabolic (dysfunction) associated fatty liver disease (MAFLD) is suggested to replace the nomenclature of NAFLD. For individuals with metabolic dysfunction, multiple NAFLD-related factors also contribute to the development and progression of MAFLD including genetics and epigenetics. The application of genome-wide association study (GWAS) and exome-wide association study (EWAS) uncovers single-nucleotide polymorphisms (SNPs) in MAFLD. In addition to the classic SNPs in PNPLA3, TM6SF2, and GCKR, some new SNPs have been found recently to contribute to the pathogenesis of liver steatosis. Epigenetic factors involving DNA methylation, histone modifications, non-coding RNAs regulations, and RNA methylation also play a critical role in MAFLD. DNA methylation is the most reported epigenetic modification. Developing a non-invasion biomarker to distinguish metabolic steatohepatitis (MASH) or liver fibrosis is ongoing. In this review, we summarized and discussed the latest progress in genetic and epigenetic factors of NAFLD/MAFLD, in order to provide potential clues for MAFLD treatment.
Collapse
Affiliation(s)
- Xiaopeng Zhu
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China
| | | | - Xin Gao
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China
| |
Collapse
|
760
|
Xiang H, Sun D, Liu X, She ZG, Chen Y. The Role of the Intestinal Microbiota in Nonalcoholic Steatohepatitis. Front Endocrinol (Lausanne) 2022; 13:812610. [PMID: 35211093 PMCID: PMC8861316 DOI: 10.3389/fendo.2022.812610] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/04/2022] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is a serious disease threatening public health, and its pathogenesis remains largely unclear. Recent scientific research has shown that intestinal microbiota and its metabolites have an important impact on the development of NASH. A balanced intestinal microbiota contributes to the maintenance of liver homeostasis, but when the intestinal microbiota is disequilibrated, it serves as a source of pathogens and molecules that lead to NASH. In this review, we mainly emphasize the key mechanisms by which the intestinal microbiota and its metabolites affect NASH. In addition, recent clinical trials and animal studies on the treatment of NASH by regulating the intestinal microbiota through prebiotics, probiotics, synbiotics and FMT have also been briefly elaborated. With the increasing understanding of interactions between the intestinal microbiota and liver, accurate and personalized detection and treatment methods for NASH are expected to be established.
Collapse
Affiliation(s)
- Hui Xiang
- Infectious Disease Department, Chongqing University Three Gorges Hospital, Chongqing, China
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- *Correspondence: Hui Xiang, ; Zhi-Gang She, ; Yonghong Chen,
| | - Dating Sun
- Department of Cardiology, Wuhan NO.1 Hospital, Wuhan, China
| | - Xin Liu
- Infectious Disease Department, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Zhi-Gang She
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- *Correspondence: Hui Xiang, ; Zhi-Gang She, ; Yonghong Chen,
| | - Yonghong Chen
- Infectious Disease Department, Chongqing University Three Gorges Hospital, Chongqing, China
- *Correspondence: Hui Xiang, ; Zhi-Gang She, ; Yonghong Chen,
| |
Collapse
|
761
|
Li T, Qu J, Xu C, Fang T, Sun B, Chen L. Exploring the common gene signatures and pathogeneses of obesity with Alzheimer's disease via transcriptome data. Front Endocrinol (Lausanne) 2022; 13:1072955. [PMID: 36568118 PMCID: PMC9780446 DOI: 10.3389/fendo.2022.1072955] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 11/22/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Obesity is a complex condition that influences several organ systems and physiologic systems. Obesity (OB) is closely linked to Alzheimer's disease (AD). However, the interrelationship between them remains unclear. The purpose of this study is to explore the key genes and potential molecular mechanisms in obesity and AD. METHODS The microarray data for OB and AD were downloaded from the Gene Expression Omnibus (GEO) database. Weighted gene correlation network analysis (WGCNA) was used to delineate the co-expression modules related to OB and AD. The shared genes existing in obesity and AD were identified through biological process analyses using the DAVID website, which then constructed the Protein-Protein Interaction (PPI) Network and selected the hub genes by Cytoscape. The results were validated in other microarray data by differential gene analysis. Moreover, the hub gene expressions were further determined in mice by qPCR. RESULTS The WGCNA identifies five modules and four modules as significant modules with OB and AD, respectively. Functional analysis of shared genes emphasized that inflammation response and mitochondrial functionality were common features in the pathophysiology of OB and AD. The results of differential gene analysis in other microarray data were extremely similar to them. Then six important hub genes were selected and identified using cytoHubba, including MMP9, PECAM1, C3AR1, IL1R1, PPARGC1α, and COQ3. Finally, we validated the hub gene expressions via qPCR. CONCLUSIONS Our work revealed the high inflammation/immune response and mitochondrial impairment in OB patients, which might be a crucial susceptibility factor for AD. Meanwhile, we identified novel gene candidates such as MMP9, PECAM1, C3AR1, IL1R1, PPARGC1α, and COQ3 that could be used as biomarkers or potential therapeutic targets for OB with AD.
Collapse
Affiliation(s)
| | | | | | | | - Bei Sun
- *Correspondence: Liming Chen, ; Bei Sun,
| | | |
Collapse
|
762
|
Asgharpour A, Sanyal AJ. Generation of a Diet-Induced Mouse Model of Nonalcoholic Fatty Liver Disease. Methods Mol Biol 2022; 2455:19-30. [PMID: 35212982 DOI: 10.1007/978-1-0716-2128-8_2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The obesity epidemic is driving the increased prevalence of nonalcoholic fatty liver disease (NAFLD) globally. The more aggressive subtype of NAFLD, nonalcoholic steatohepatitis (NASH), can lead to progressive disease and ultimately lead to cirrhosis, liver cancer, and death. There are many unmet needs in the field of NAFLD including understanding of molecular mechanisms driving disease, natural history, risk for liver cancer, and most importantly FDA approved therapeutics. Animal models serve as a tool to aid in answering some of these questions. Here, we describe the diet-induced animal model of NAFLD (DIAMOND), a mouse model with many characteristics that mimic human NASH.
Collapse
Affiliation(s)
- Amon Asgharpour
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA.
| | - Arun J Sanyal
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
763
|
Gao W, Li Z, Chu H, Yuan H, Hu L, Yao L, Zhang L, Wang W, Lin R, Yang L. Ursodeoxycholic Acid in Liver Cirrhosis: A Chinese Perspective. PHARMACOTHERAPY FOR LIVER CIRRHOSIS AND ITS COMPLICATIONS 2022:81-111. [DOI: 10.1007/978-981-19-2615-0_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
764
|
Gu W, Hortlik H, Erasmus HP, Schaaf L, Zeleke Y, Uschner FE, Ferstl P, Schulz M, Peiffer KH, Queck A, Sauerbruch T, Brol MJ, Rohde G, Sanchez C, Moreau R, Arroyo V, Zeuzem S, Welsch C, Trebicka J. Trends and the course of liver cirrhosis and its complications in Germany: Nationwide population-based study (2005 to 2018). THE LANCET REGIONAL HEALTH. EUROPE 2022; 12:100240. [PMID: 34901909 PMCID: PMC8640738 DOI: 10.1016/j.lanepe.2021.100240] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Cirrhosis is known to have a high prevalence and mortality worldwide. However, in Europe, the epidemiology of cirrhosis is possibly undergoing demographic changes, and etiologies may have changed due to improvements in standard of care. The aim of this population-based study was to analyze the trends and the course of liver cirrhosis and its complications in recent years in Germany. METHODS We analyzed the data of all hospital admissions in Germany within diagnosis-related groups from 2005 to 2018. The diagnostic records of cirrhosis and other categories of diseases were based on ICD-10-GM codes. The primary outcome measurement was in-hospital mortality. Trends were analyzed through Poisson regression of annual number of admissions. The impact of cirrhosis on overall in-hospital mortality were assessed through the multivariate multilevel logistic regression model adjusted for age, sex, and comorbidities. FINDINGS Of the 248,085,936 admissions recorded between 2005 and 2018, a total of 2,302,171(0•94%) were admitted with the diagnosis of cirrhosis, mainly as a comorbidity. Compared with other chronic diseases, patients admitted with cirrhosis were younger, mainly male and had the highest in-hospital mortality rate. Diagnosis of cirrhosis was an independent risk factor of in-hospital mortality with the highest odds ratio (OR:6•2[95%CI:6.1-6•3]) among all diagnoses. The prevalence of non-alcoholic fatty liver disease has increased four times from 2005 to 2018, while alcoholic cirrhosis is 20 times than other etiologies. Bleeding was found to be decreasing over time, but ascites remained the most common complication and was increasing. INTERPRETATION This nationwide study demonstrates that cirrhosis represents a considerable healthcare burden, as shown by the increasing in-hospital mortality, also in combination with other chronic diseases. Alcohol-related cirrhosis and complications are on the rise. More resources and better management strategies are warranted. FUNDING The funders had no influence on this study.
Collapse
Key Words
- ALD, alcoholic liver diseases
- DAA, direct-acting antiviral
- DALYs, disability-adjusted life years
- HBV, hepatitis B virus
- HCV, hepatitis C virus
- ICD, International Classification of Diseases
- NAFLD, non-alcoholic fatty liver disease
- NASH, non-alcoholic steatohepatitis
- OPS, operation and procedure key system
- YLDs, years lived with disability
- YLLs, years of life lost
- cirrhosis
- hospital admission
- male
- mortality
Collapse
Affiliation(s)
- Wenyi Gu
- Department of Internal Medicine I, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Hannah Hortlik
- Department of Internal Medicine I, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Hans-Peter Erasmus
- Department of Internal Medicine I, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Louisa Schaaf
- Department of Internal Medicine I, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Yasmin Zeleke
- Department of Internal Medicine I, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Frank E. Uschner
- Department of Internal Medicine I, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Philip Ferstl
- Department of Internal Medicine I, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Martin Schulz
- Department of Internal Medicine I, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Kai-Henrik Peiffer
- Department of Internal Medicine I, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Alexander Queck
- Department of Internal Medicine I, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Tilman Sauerbruch
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
| | - Maximilian Joseph Brol
- Department of Internal Medicine I, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Gernot Rohde
- University Hospital Frankfurt, Department of Respiratory Medicine and Allergology, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Cristina Sanchez
- European Foundation for Study of Chronic Liver Failure, Barcelona, Spain
| | - Richard Moreau
- European Foundation for Study of Chronic Liver Failure, Barcelona, Spain
- APHP, Hôpital Beaujon, Service d'Hépatologie, Clichy, France; Inserm, Université de Paris, Centre de Recherche sur L´Inflammation, Paris, France
| | - Vicente Arroyo
- European Foundation for Study of Chronic Liver Failure, Barcelona, Spain
| | - Stefan Zeuzem
- Department of Internal Medicine I, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Christoph Welsch
- Department of Internal Medicine I, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Jonel Trebicka
- Department of Internal Medicine I, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
- European Foundation for Study of Chronic Liver Failure, Barcelona, Spain
| |
Collapse
|
765
|
Ling Q, Chen J, Liu X, Xu Y, Ma J, Yu P, Zheng K, Liu F, Luo J. The triglyceride and glucose index and risk of nonalcoholic fatty liver disease: A dose-response meta-analysis. Front Endocrinol (Lausanne) 2022; 13:1043169. [PMID: 36743937 PMCID: PMC9892833 DOI: 10.3389/fendo.2022.1043169] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 12/28/2022] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND The triglyceride and glucose (TyG) index is associated with the risk of nonalcoholic fatty liver disease (NAFLD), but the dose-response relationship between them is still unknown. We conducted a comprehensive meta-analysis to study the dose-response association between the TyG index and the risk of NAFLD. METHODS We systematically searched the Cochrane Library, PubMed, and Embase databases until July 2022 for relevant studies. The robust error meta-regression method was used to investigate the dose-response association between the TyG index and NAFLD. Summary relative risks (ORs) and 95% CIs were estimated by using a random-effects model. RESULTS A total of 4 cohort and 8 cross-sectional studies were included, with 28,788 NAFLD cases among the 105,365 participants. A positive association for the risk of NAFLD was observed for each additional unit of the TyG index with a linear association (p=0.82), and the summary OR was 2.84 (95% CI, 2.01-4.01). In the subgroup analyses, a stronger association of the TyG index with NAFLD was shown in females than in males (men: OR=2.97, 95% CI 2.55-3.46, women: OR=4.80, 95% CI 3.90-5.90, Psubgroup<0.001). CONCLUSION The TyG index may be a novel independent risk factor for NAFLD beyond traditional risk factors. SYSTEMATIC REVIEW REGISTRATION https://www.crd.york.ac.uk/prospero, identifier (CRD42022347813).
Collapse
Affiliation(s)
- Qin Ling
- Department of Cardiology, the Affiliated Ganzhou Hospital of Nanchang University, Jiangxi, China
- The Second Clinical Medical College of Nanchang University, Jiangxi, China
| | - Jiawei Chen
- Department of Cardiology, the Affiliated Ganzhou Hospital of Nanchang University, Jiangxi, China
- The Second Clinical Medical College of Nanchang University, Jiangxi, China
| | - Xiao Liu
- Department of Cardiology, the Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yi Xu
- Department of Cardiology, the Affiliated Ganzhou Hospital of Nanchang University, Jiangxi, China
- The Second Clinical Medical College of Nanchang University, Jiangxi, China
| | - Jianyong Ma
- Department of Pharmacology and Systems Physiology, University of Cinnati College of Medicine, Cincinnati, OH, United States
| | - Peng Yu
- Department of Endocrine, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Kai Zheng
- Medical Care Strategic Customer Department, China Merchants Bank Shenzhen Branch, Shenzhen, China
| | - Fuwei Liu
- Department of Cardiology, the Affiliated Ganzhou Hospital of Nanchang University, Jiangxi, China
- *Correspondence: Jun Luo, ; Fuwei Liu,
| | - Jun Luo
- Department of Cardiology, the Affiliated Ganzhou Hospital of Nanchang University, Jiangxi, China
- *Correspondence: Jun Luo, ; Fuwei Liu,
| |
Collapse
|
766
|
Hepatic macrophage targeted siRNA lipid nanoparticles treat non-alcoholic steatohepatitis. J Control Release 2022; 343:175-186. [DOI: 10.1016/j.jconrel.2022.01.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 12/12/2021] [Accepted: 01/23/2022] [Indexed: 12/12/2022]
|
767
|
Zhang S, Huang YP, Li J, Wang WH, Zhang MY, Wang XC, Lin JN, Li CJ. The Visceral-Fat-Area-to-Hip-Circumference Ratio as a Predictor for Insulin Resistance in a Chinese Population with Type 2 Diabetes. Obes Facts 2022; 15:621-628. [PMID: 35724630 PMCID: PMC9421683 DOI: 10.1159/000525545] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 03/03/2022] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Adipose tissue deposited on the viscera is the main culprit in the development of insulin resistance (IR) and cardiometabolic diseases, whereas subcutaneous adipose tissue may have a protective role. This study aimed to propose a new predictive index - the visceral-fat area (VFA)-to-hip-circumference ratio (VHR) and explore its efficacy for prediction of IR in a Chinese population with type 2 diabetes mellitus. METHODS A total of 643 patients with newly diagnosed diabetes were enrolled in this study. Body composition, anthropometrical, and biochemical measurements were performed. IR was defined as homeostatic model assessment of IR (HOMA-IR) > 2.69. The association between VHR and IR was analyzed. RESULTS Regardless of gender, subjects in the IR group had higher VHR, body mass index (BMI), VFA, body fat percentage, systolic blood pressure, diastolic blood pressure (DBP), fasting blood glucose, fasting insulin, triglyceride (TG), uric acid (UA), homocysteine, and aminotransferases than those in the non-IR group. The other concomitant metabolic disorders were more common in the IR group. Further analysis showed that with the increase of VHR, the levels of HOMA-IR, BMI, VFA, DBP, TG, UA and the prevalence of nonalcoholic fatty-liver disease, hypertension, and hyperuricemia increased continuously (p trend <0.01). The linear trend test showed that VHR and IR remained closely correlated after adjusting for possible confounders (p trend <0.05). The receiver operating characteristic curve analysis showed that the area under the curve was 0.69, and the optimal cutoff of VHR was 0.89 (sensitivity 79.3%, specificity 61.5%). CONCLUSION VHR was positively associated with IR regardless of gender, and it might be a reliable predictor for IR.
Collapse
Affiliation(s)
- Shi Zhang
- Department of Endocrinology, Health Management Center, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, China,
| | - Ya-Ping Huang
- Department of Endocrinology, Health Management Center, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, China
- Tianjin Medical University, Tianjin, China
| | - Jing Li
- Department of Endocrinology, Health Management Center, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, China
| | - Wen-Hong Wang
- Department of Endocrinology, Health Management Center, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, China
| | | | - Xin-Cheng Wang
- Department of Endocrinology, Health Management Center, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, China
| | - Jing-Na Lin
- Department of Endocrinology, Health Management Center, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, China
| | - Chun-Jun Li
- Department of Endocrinology, Health Management Center, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, China
| |
Collapse
|
768
|
Liu Q, Li H, He W, Zhao Q, Huang C, Wang Q, Zheng Z, Zhang X, Shi X, Li X. Role of aerobic exercise in ameliorating NASH: Insights into the hepatic thyroid hormone signaling and circulating thyroid hormones. Front Endocrinol (Lausanne) 2022; 13:1075986. [PMID: 36605939 PMCID: PMC9807753 DOI: 10.3389/fendo.2022.1075986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022] Open
Abstract
AIM Triiodothyronine (T3) administration significantly eliminates hepatic steatosis and also has a therapeutic effect on non-alcoholic steatohepatitis (NASH). However, the potential mechanism by which T3-mediated exercise improves NASH is unknow. This study aimed to explore the effect of aerobic exercise on liver injury in NASH. METHODS Aerobic exercise was conducted to explore the effects of exercise on liver injury in NASH model induced by Atherosclerotic (Ath) diet. Biochemical evaluations, histological staining and real-time PCR were first applied to confirm the amelioration effects of exercise on NASH. RNA-sequencing (RNA-seq) analysis for livers of each group were further used to identify the underlying mechanisms of aerobic exercise. Bioinformatics methods were used to explore the key functional pathways involved in the improvement of liver tissue in NASH mice by aerobic exercise. RESULTS Aerobic exercise improved hepatic steatosis, lobular inflammation and fibrosis in NASH mice. multiple inflammation-related pathways were significantly enriched in the liver of NASH group and improved by aerobic exercise. The results of gene set variation analysis (GSVA) showed a higher enrichment score of T3 response signature in NASH mice with exercise. Increased Dio1 expression in the liver of NASH with exercise mice and increased circulating FT3 and FT4 levels upon aerobic exercise were confirmed. CONCLUSIONS We found that aerobic exercise could significantly reduce hepatic lipid accumulation, inflammatory infiltration and fibrosis progression in the liver of NASH mice. Hepatic thyroid hormone signaling activation and circulating thyroid hormones is potentially involved in the amelioration effect of aerobatic exercise on NASH progression.
Collapse
Affiliation(s)
- Qiuhong Liu
- School of Medicine, Xiamen University, Xiamen, China
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Han Li
- School of Medicine, Xiamen University, Xiamen, China
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Weiwei He
- School of Medicine, Xiamen University, Xiamen, China
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Qing Zhao
- School of Medicine, Xiamen University, Xiamen, China
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Caoxin Huang
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Qingxuan Wang
- School of Medicine, Xiamen University, Xiamen, China
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Zeyu Zheng
- School of Medicine, Xiamen University, Xiamen, China
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xiaofang Zhang
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xiulin Shi
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- *Correspondence: Xuejun Li, ; Xiulin Shi,
| | - Xuejun Li
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- *Correspondence: Xuejun Li, ; Xiulin Shi,
| |
Collapse
|
769
|
Tong J, Cong L, Jia Y, He BL, Guo Y, He J, Li D, Zou B, Li J. Follistatin Alleviates Hepatic Steatosis in NAFLD via the mTOR Dependent Pathway. Diabetes Metab Syndr Obes 2022; 15:3285-3301. [PMID: 36325432 PMCID: PMC9621035 DOI: 10.2147/dmso.s380053] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 10/07/2022] [Indexed: 11/13/2022] Open
Abstract
PURPOSE In this study, we aimed to investigate the effect of follistatin (FST) on hepatic steatosis in NAFLD and the underlying mechanism, which has rarely been reported before. METHODS Liver samples from NAFLD patients and normal liver samples (from liver donors) were collected to investigate hepatic FST expression in humans. Additionally, human liver cells (LO2) were treated with free fatty acid (FFA) to induce lipid accumulation. Furthermore, lentivirus with FST overexpression or knockdown vectors were used to generate stable cell lines, which were subsequently treated with FFA or FFA and rapamycin. In the animal experiments, male C57BL/6J mice were fed with a high-fat diet (HFD) to induce NAFLD, after which the adeno-associated virus (AAV) gene vectors for FST overexpression were administered. In both cell culture and mice, we evaluated morphological changes and the protein expression of sterol regulatory element-binding protein1 (SREBP1), acetyl-CoA carboxylase1 (ACC1), carbohydrate-responsive element-binding protein (ChREBP), fatty acid synthase (FASN), and Akt/mTOR signaling. The body weight and serum parameters of the mice were also measured. RESULTS Hepatic FST expression level was higher in NAFLD patients compared to normal samples. In LO2 cells, FST overexpression alleviated lipid accumulation and lipogenesis, whereas FST knockdown aggravated hepatic steatosis. FST could regulate Akt/mTOR signaling, and the mTOR inhibitor rapamycin abolished the effect of FST knockdown on hepatic de novo lipogenesis (DNL). Furthermore, FST expression was increased in HFD mice compared to the corresponding controls. FST overexpression in mice reduced body weight gain, hyperlipidemia, hepatic DNL, and suppressed Akt/mTOR signaling. CONCLUSION Hepatic FST expression increases in NAFLD and plays a protective role in hepatic steatosis. FST overexpression gene therapy alleviates hepatic steatosis via the mTOR pathway.Therefore, gene therapy for FST is a promising treatment in NAFLD.
Collapse
Affiliation(s)
- Junlu Tong
- Department of Endocrinology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, People’s Republic of China
- Department of Central Laboratory, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, People’s Republic of China
| | - Li Cong
- Department of Endocrinology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, People’s Republic of China
| | - Yingbin Jia
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, People’s Republic of China
| | - Bai-Liang He
- Guangdong Provincial Key Laboratory of Biomedical Imaging, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, People’s Republic of China
| | - Yifan Guo
- Department of Endocrinology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, People’s Republic of China
| | - Jianzhong He
- Department of Pathology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, People’s Republic of China
| | - Decheng Li
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, People’s Republic of China
| | - Baojia Zou
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, People’s Republic of China
| | - Jian Li
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, People’s Republic of China
- Correspondence: Jian Li; Baojia Zou, Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, People’s Republic of China, Tel +86-756-252-8781, Email ;
| |
Collapse
|
770
|
Seeberg KA, Borgeraas H, Hofsø D, Småstuen MC, Kvan NP, Grimnes JO, Lindberg M, Fatima F, Seeberg LT, Sandbu R, Hjelmesæth J, Hertel JK. Gastric Bypass Versus Sleeve Gastrectomy in Type 2 Diabetes: Effects on Hepatic Steatosis and Fibrosis : A Randomized Controlled Trial. Ann Intern Med 2022; 175:74-83. [PMID: 34843380 DOI: 10.7326/m21-1962] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Weight loss improves fatty liver disease. No randomized trial has compared the effects of sleeve gastrectomy (SG) and Roux-en-Y gastric bypass (RYGB) on liver fat content and fibrosis. OBJECTIVE To compare the 1-year effects of SG and RYGB on hepatic steatosis and fibrosis. DESIGN Single-center, randomized, controlled trial (Oseberg [ObesitySurgery in Tønsberg]). (ClinicalTrials.gov: NCT01778738). SETTING Tertiary care obesity center in Norway. PARTICIPANTS 100 patients (65% female; mean age, 47.5 years; mean body mass index, 42 kg/m2) with type 2 diabetes mellitus (T2DM). INTERVENTION From January 2013 to February 2018, patients were randomly assigned (1:1 ratio) to SG or RYGB. MEASUREMENTS The primary outcome was remission of T2DM (previously published). Predefined secondary outcomes in the present study were hepatic steatosis and fibrosis assessed by magnetic resonance imaging (liver fat fraction), enhanced liver fibrosis (ELF) test, noninvasive indices, and liver enzymes. RESULTS Liver fat fraction declined similarly after SG (-19.7% [95% CI, -22.5% to -16.9%]) and RYGB (-21.5% [CI, -24.3% to -18.6%]) from surgery to 1-year follow-up, and almost all patients (SG, 94%; RYGB, 100%) had no or low-grade steatosis at 1 year. The ELF score category remained stable in 77% of patients, but 18% experienced worsening of fibrosis at 1 year, with no substantial between-group difference. LIMITATIONS Single-center study, short follow-up time, and lack of power for secondary outcomes. CONCLUSION With an almost complete clearance of liver fat 1 year after surgery, RYGB and SG were both highly effective in reducing hepatic steatosis. Bariatric surgery had less influence on degree of fibrosis in the short term, but assessment of long-term progression is warranted. PRIMARY FUNDING SOURCE Vestfold Hospital Trust and the South-Eastern Norway Regional Health Authority.
Collapse
Affiliation(s)
- Kathrine Aglen Seeberg
- Morbid Obesity Center and Department of Medicine, Vestfold Hospital Trust, Tønsberg, and Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway (K.A.S.)
| | - Heidi Borgeraas
- Morbid Obesity Center, Vestfold Hospital Trust, Tønsberg, Norway (H.B., J.K.H.)
| | - Dag Hofsø
- Morbid Obesity Center and Department of Medicine, Vestfold Hospital Trust, Tønsberg, Norway (D.H.)
| | - Milada Cvancarova Småstuen
- Morbid Obesity Center, Vestfold Hospital Trust, Tønsberg, and Department of Nursing and Health Promotion, Oslo Metropolitan University, Oslo, Norway (M.C.S.)
| | - Nils Petter Kvan
- Department of Radiology, Vestfold Hospital Trust, Tønsberg, Norway (N.P.K., J.O.G.)
| | - John Olav Grimnes
- Department of Radiology, Vestfold Hospital Trust, Tønsberg, Norway (N.P.K., J.O.G.)
| | - Morten Lindberg
- Department of Laboratory Medicine, Vestfold Hospital Trust, Tønsberg, Norway (M.L.)
| | - Farhat Fatima
- Morbid Obesity Center, Vestfold Hospital Trust, Tønsberg, and Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway (F.F.)
| | | | - Rune Sandbu
- Morbid Obesity Center and Department of Surgery, Vestfold Hospital Trust, Tønsberg, Norway (R.S.)
| | - Jøran Hjelmesæth
- Morbid Obesity Center, Vestfold Hospital Trust, Tønsberg, and Department of Endocrinology, Morbid Obesity and Preventive Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway (J.H.)
| | | |
Collapse
|
771
|
Lv S, Zhang Z, Su X, Li W, Wang X, Pan B, Li H, Zhang H, Wang Y. Qingrequzhuo capsule alleviated methionine and choline deficient diet-induced nonalcoholic steatohepatitis in mice through regulating gut microbiota, enhancing gut tight junction and inhibiting the activation of TLR4/NF-κB signaling pathway. Front Endocrinol (Lausanne) 2022; 13:1106875. [PMID: 36743916 PMCID: PMC9892721 DOI: 10.3389/fendo.2022.1106875] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 12/29/2022] [Indexed: 01/20/2023] Open
Abstract
Qingrequzhuo capsule (QRQZ), composed of Morus alba L., Coptis chinensis Franch., Anemarrhena asphodeloides Bunge, Alisma plantago-aquatica subsp. orientale (Sam.) Sam., Citrus × aurantium L., Carthamus tinctorius L., Rheum palmatum L., Smilax glabra Roxb., Dioscorea oppositifolia L., Cyathula officinalis K.C.Kuan, has been used to treat nonalcoholic steatohepatitis (NASH) in clinic. However, the mechanism of QRQZ on NASH remains unclear. Recent studies have found that the dysfunction of gut microbiota could impair the gut barrier and induce the activation of TLR4/NF-kB signaling pathway, and further contribute to the inflammatory response in NASH. Modulating the gut microbiota to reduce inflammation could prevent the progression of NASH. In this study, a mouse model of NASH was generated by methionine and choline deficient diet (MCD) and treated with QRQZ. First, we evaluated the therapeutic effects of QRQZ on liver injury and inflammation in the NASH mice. Second, the changes in the gut microbiota diversity and abundance in each group of mice were measured through 16S rRNA sequencing. Finally, the effects of QRQZ on gut mucosal permeability, endotoxemia, and liver TLR4/NF-kB signaling pathway levels were examined. Our results showed that QRQZ significantly reduced the lipid accumulation in liver and the liver injury in NASH mice. In addition, QRQZ treatment decreased the levels of inflammatory cytokines in liver. 16S rRNA sequencing showed that QRQZ affected the diversity of gut microbiota and a f f e c t e d t h e r e l a t i v e a b u n d a n c e s o f D u b o s i e l l a , Lachnospiraceae_NK4A136_group, and Blautiain NASH mice. Besides, QRQZ could increase the expression of tight junction proteins (zonula occludens-1 and occludin) in gut and decrease the lipopolysaccharide (LPS) level in serum. Western blot results also showed that QRQZ treatment decreased the protein expression ofTLR4, MyD88 and the phosphorylation of IkB and NF-kBp65 and qPCR results showed that QRQZ treatment down-regulated the gene expression of interleukin (IL)-1b, IL-6, and tumor necrosis factor (TNF)-a in liver. In conclusion, our study demonstrated that QRQZ could reduce the lipid accumulation and inflammatory response in NASH model mice. The mechanisms of QRQZ on NASH were associated with modulating gut microbiota, thereby inducing the tight junction of gut barrier, reducing the endotoxemia and inhibiting the activation of TLR4/NFkB signaling pathway in liver.
Collapse
Affiliation(s)
- Shuquan Lv
- Department of Endocrinology, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, China
| | - Zhongyong Zhang
- Department of Endocrinology, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, China
| | - Xiuhai Su
- Department of Endocrinology, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, China
| | - Wendong Li
- Department of Endocrinology, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, China
| | - Xiaoyun Wang
- Department of Endocrinology, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, China
| | - Baochao Pan
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Hanzhou Li
- Graduate School, Chengde Medical University, Chengde, China
| | - Hui Zhang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yuansong Wang
- Department of Endocrinology, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, China
- *Correspondence: Yuansong Wang,
| |
Collapse
|
772
|
Vieira Barbosa J, Milligan S, Frick A, Broestl J, Younossi Z, Afdhal NH, Lai M. Fibrosis-4 Index as an Independent Predictor of Mortality and Liver-Related Outcomes in NAFLD. Hepatol Commun 2021; 6:765-779. [PMID: 34970870 PMCID: PMC8948572 DOI: 10.1002/hep4.1841] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 07/30/2021] [Accepted: 09/21/2021] [Indexed: 01/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease worldwide, and its prevalence continues to rise. Fibrosis-4 index (FIB-4) has been shown to be a prognostic marker of liver-related outcomes in patients with NAFLD. We analyzed data from TriNetX global federated research network, combining data on 30 million patients. Patients were categorized into three diagnostic groups: NAFLD, nonalcoholic steatohepatitis (NASH), and at risk of NASH. Primary outcome was all-cause mortality, and secondary outcomes included progression to NASH, development of cirrhosis, end-stage liver disease, hepatocellular carcinoma (HCC), and liver transplantation. A total of 442,277 subjects (1.5% of the cohort) were assessed, and 81,108 were retained for analysis. Median follow-up was 34.8 months (interquartile range 12.2). FIB-4 was < 1.3 in 52.3% patients and ≥ 2.67 in 11.4% patients. In multivariate analysis, FIB-4 ≥ 2.67 was significantly and independently associated with all-cause mortality (hazard ratio [HR] 2.49, 95% confidence interval [CI] 2.20-2.82, P < 0.001) as well as with progression to NASH (HR 5.78, 95% CI 4.72-7.07, P < 0.001), cirrhosis (HR 2.04, 95% CI 1.86-2.24, P < 0.001), end-stage liver disease (HR 1.86, 95% CI 1.68-2.05, P < 0.001), HCC (HR 3.66, 95% CI 2.71-4.94, P < 0.001), and liver transplantation (HR 7.98, 95% CI 4.62-13.79, P < 0.001). Conclusion: In a real-world nationwide database, FIB-4 ≥ 2.67 was a strong predictor of both all-cause mortality and liver-related adverse outcomes independently of the baseline diagnostic group and common risk factors. Our findings indicate that FIB-4 could play a role as a risk-stratification tool for a population health approach. Significant underdiagnosis of both NAFLD/NASH and NASH cirrhosis in electronic medical records was observed.
Collapse
Affiliation(s)
- Joana Vieira Barbosa
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.,Division of Gastroenterology and Hepatology, Lausanne University Hospital and University of Lausanne, Switzerland
| | | | | | | | - Zobair Younossi
- Center for Liver Diseases, Department of Medicine, Inova Fairfax Hospital, Falls Church, VA, USA.,Betty and Guy Beatty Center for Integrated Research, Inova Health System, Falls Church, VA, USA
| | - Nezam H Afdhal
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Michelle Lai
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
773
|
Cao S, Yang X, Zhang Z, Wu J, Chi B, Chen H, Yu J, Feng S, Xu Y, Li J, Zhang Y, Wang X, Wang Y. Discovery of a tricyclic farnesoid X receptor agonist HEC96719, a clinical candidate for treatment of non-alcoholic steatohepatitis. Eur J Med Chem 2021; 230:114089. [PMID: 34998040 DOI: 10.1016/j.ejmech.2021.114089] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/24/2021] [Accepted: 12/26/2021] [Indexed: 11/04/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is becoming the most predominant burden of chronic liver disease worldwide. Non-alcoholic steatohepatitis (NASH), the progressive form of NAFLD, can develop into cirrhosis and hepatocellular cancer. Unfortunately, current options for therapeutic treatment of NASH are very limited. Among multiple pathways in NASH, farnesoid X receptor (FXR), a nuclear bile acid receptor, is well-recognized as an important effective target. Here we report the synthesis and characterization of compound HEC96719 a novel tricyclic FXR agonist based on a prior high-affinity nonsteroidal molecule GW4064. HEC96719 exhibits excellent potency superior to GW4064 and obeticholic acid in in vitro and in vivo assays of FXR activation. It also shows higher FXR selectivity and more favorable tissue distribution dominantly in liver and intestine. Preclinical data on pharmacokinetic properties, efficacy, and safety profiles overall indicate that HEC96719 is a promising drug candidate for NASH treatment.
Collapse
Affiliation(s)
- Shengtian Cao
- Southern Medical University Biomedical Research Center, Guangdong Provincial Research Center for Liver Fibrosis, Southern Medical University, Guangzhou, Guangdong, China; Sunshine Lake Pharma Co Ltd, HEC Pharm Group, HEC Research and Development Center, Dongguan, Guangdong, China
| | - Xinye Yang
- Sunshine Lake Pharma Co Ltd, HEC Pharm Group, HEC Research and Development Center, Dongguan, Guangdong, China
| | - Zheng Zhang
- Sunshine Lake Pharma Co Ltd, HEC Pharm Group, HEC Research and Development Center, Dongguan, Guangdong, China
| | - Junwen Wu
- Sunshine Lake Pharma Co Ltd, HEC Pharm Group, HEC Research and Development Center, Dongguan, Guangdong, China
| | - Bo Chi
- Sunshine Lake Pharma Co Ltd, HEC Pharm Group, HEC Research and Development Center, Dongguan, Guangdong, China
| | - Hong Chen
- Sunshine Lake Pharma Co Ltd, HEC Pharm Group, HEC Research and Development Center, Dongguan, Guangdong, China
| | - Jianghong Yu
- Sunshine Lake Pharma Co Ltd, HEC Pharm Group, HEC Research and Development Center, Dongguan, Guangdong, China
| | - Shanshan Feng
- Sunshine Lake Pharma Co Ltd, HEC Pharm Group, HEC Research and Development Center, Dongguan, Guangdong, China
| | - Yulin Xu
- Sunshine Lake Pharma Co Ltd, HEC Pharm Group, HEC Research and Development Center, Dongguan, Guangdong, China
| | - Jing Li
- Sunshine Lake Pharma Co Ltd, HEC Pharm Group, HEC Research and Development Center, Dongguan, Guangdong, China
| | - Yingjun Zhang
- Sunshine Lake Pharma Co Ltd, HEC Pharm Group, HEC Research and Development Center, Dongguan, Guangdong, China
| | - Xiaojun Wang
- Sunshine Lake Pharma Co Ltd, HEC Pharm Group, HEC Research and Development Center, Dongguan, Guangdong, China.
| | - Yan Wang
- Southern Medical University Biomedical Research Center, Guangdong Provincial Research Center for Liver Fibrosis, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
774
|
Association between Non-Alcoholic Fatty Liver Disease and Mediterranean Lifestyle: A Systematic Review. Nutrients 2021; 14:nu14010049. [PMID: 35010923 PMCID: PMC8746321 DOI: 10.3390/nu14010049] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 12/16/2021] [Accepted: 12/21/2021] [Indexed: 02/07/2023] Open
Abstract
Background and Aims: Non-alcoholic fatty liver disease (NAFLD) is an excessive accumulation of fat in the liver without alcohol abuse. It is linked to metabolic syndrome (MetS) and no pharmacological treatment exists. This systematic review aims to assess evidence about the effect of Mediterranean lifestyle on the prevention and reversion of NAFLD. Methods: A systematic literature search was performed in MEDLINE via Pubmed. MeSH terms used were: non-alcoholic fatty liver disease [MeSH Major Topic] AND metabolic syndrome [MeSH Term] AND (Diet, Mediterranean [MeSH Term]) OR (Exercise [MeSH Term]). (PROSPERO ID: 2021 CRD42021289495). Results: Thirteen articles were selected and divided into two categories (four focused on Mediterranean diet and NAFLD and nine focused on Mediterranean diet, physical activity, and NAFLD). Information of clinical endpoints was based on NAFLD, as well as MetS, body mass index, fasting glycemia, obesity, cholesterol, triglycerides, transaminases, albuminuria, and hepatic steatosis, among others. All studies found beneficial associations between the clinical parameters of NAFLD/MetS and following a Mediterranean diet and regular physical activity. Conclusions: An effective treatment that prevents, and even reverses, NAFLD is to adapt lifestyle to the Mediterranean one, based on a Mediterranean diet and regular physical activity.
Collapse
|
775
|
Balak DMW, Piaserico S, Kasujee I. Non-Alcoholic Fatty Liver Disease (NAFLD) in Patients with Psoriasis: A Review of the Hepatic Effects of Systemic Therapies. PSORIASIS (AUCKLAND, N.Z.) 2021; 11:151-168. [PMID: 34909410 PMCID: PMC8665778 DOI: 10.2147/ptt.s342911] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 11/09/2021] [Indexed: 12/12/2022]
Abstract
There is increasing interest in the association between psoriasis and non-alcoholic fatty liver disease (NAFLD), which is a prevalent liver disease characterized by excessive fat storage and inflammation that can progress to fibrosis and cancer. Patients with psoriasis have a two-fold higher risk to develop NAFLD and a higher risk to progress to more severe liver disease. Psoriasis and NAFLD share common risk factors such as smoking, alcohol consumption, and the presence of metabolic syndrome and its component disorders. In addition, both psoriasis and NAFLD hinge upon a systemic low-grade inflammation that can lead to a vicious cycle of progressive liver damage in NAFLD as well as worsening of the underlying psoriasis. Other important shared pathophysiological pathways include peripheral insulin resistance and oxidative stress. NAFLD should receive clinical awareness as important comorbidity in psoriasis. In this review, we assess the recent literature on the epidemiological and pathophysiological relationship of psoriasis and NAFLD, discuss the clinical implications of NAFLD in psoriasis patients, and summarize the hepatotoxic and hepatoprotective potential of systemic psoriasis therapies.
Collapse
Affiliation(s)
- Deepak M W Balak
- Department of Dermatology, LangeLand Ziekenhuis, Zoetermeer, the Netherlands.,Department of Dermatology, Ghent University Hospital, Ghent, Belgium
| | - Stefano Piaserico
- Dermatology Unit, Department of Medicine, University of Padova, Padova, Italy
| | | |
Collapse
|
776
|
Zhang XJ, Ji YX, Cheng X, Cheng Y, Yang H, Wang J, Zhao LP, Huang YP, Sun D, Xiang H, Shen LJ, Li PL, Ma JP, Tian RF, Yang J, Yao X, Xu H, Liao R, Xiao L, Zhang P, Zhang X, Zhao GN, Wang X, Hu ML, Tian S, Wan J, Cai J, Ma X, Xu Q, Wang Y, Touyz RM, Liu PP, Loomba R, She ZG, Li H. A small molecule targeting ALOX12-ACC1 ameliorates nonalcoholic steatohepatitis in mice and macaques. Sci Transl Med 2021; 13:eabg8116. [PMID: 34910548 DOI: 10.1126/scitranslmed.abg8116] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Xiao-Jing Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Institute of Model Animal of Wuhan University, Wuhan 430071, China.,School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Yan-Xiao Ji
- Institute of Model Animal of Wuhan University, Wuhan 430071, China.,School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Xu Cheng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Yanjie Cheng
- Institute of Model Animal of Wuhan University, Wuhan 430071, China.,Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Hailong Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Junyong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Ling-Ping Zhao
- Institute of Model Animal of Wuhan University, Wuhan 430071, China.,School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Yong-Ping Huang
- Institute of Model Animal of Wuhan University, Wuhan 430071, China.,College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Dating Sun
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Hui Xiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Li-Jun Shen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Peng-Long Li
- Institute of Model Animal of Wuhan University, Wuhan 430071, China.,School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Jun-Peng Ma
- Institute of Model Animal of Wuhan University, Wuhan 430071, China.,School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Rui-Feng Tian
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Juan Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Xinxin Yao
- Institute of Model Animal of Wuhan University, Wuhan 430071, China.,School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Haibo Xu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Rufang Liao
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Li Xiao
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Peng Zhang
- Institute of Model Animal of Wuhan University, Wuhan 430071, China.,School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Xin Zhang
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Guang-Nian Zhao
- Institute of Model Animal of Wuhan University, Wuhan 430071, China.,Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Man-Li Hu
- Institute of Model Animal of Wuhan University, Wuhan 430071, China.,Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Song Tian
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Juan Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Jingjing Cai
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Xinliang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19004, USA
| | - Qingbo Xu
- Centre for Clinic Pharmacology, The William Harvey Research Institute, Queen Mary University of London, London SE5 9NU, UK
| | - Yibin Wang
- Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Rhian M Touyz
- British Heart Foundation Chair in Cardiovascular Medicine, and Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Peter P Liu
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ontario K1Y 4W7, Canada
| | - Rohit Loomba
- NAFLD Research Center, Division of Gastroenterology and Epidemiology, University of California, San Diego, San Diego, CA 92093, USA
| | - Zhi-Gang She
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Institute of Model Animal of Wuhan University, Wuhan 430071, China.,School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China.,Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| |
Collapse
|
777
|
Systematic Review with Meta-Analysis: Diagnostic Accuracy of Pro-C3 for Hepatic Fibrosis in Patients with Non-Alcoholic Fatty Liver Disease. Biomedicines 2021; 9:biomedicines9121920. [PMID: 34944736 PMCID: PMC8698886 DOI: 10.3390/biomedicines9121920] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/03/2021] [Accepted: 12/10/2021] [Indexed: 12/26/2022] Open
Abstract
The prevalence and severity of non-alcoholic fatty liver disease (NAFLD) is increasing, yet adequately validated tests for care paths are limited and non-invasive markers of disease progression are urgently needed. The aim of this work was to summarize the performance of Pro-C3, a biomarker of active fibrogenesis, in detecting significant fibrosis (F ≥ 2), advanced fibrosis (F ≥ 3), cirrhosis (F4) and non-alcoholic steatohepatitis (NASH) in patients with NAFLD. A sensitive search of five databases was performed in July 2021. Studies reporting Pro-C3 measurements and liver histology in adults with NAFLD without co-existing liver diseases were eligible. Meta-analysis was conducted by applying a bivariate random effects model to produce summary estimates of Pro-C3 accuracy. From 35 evaluated reports, eight studies met our inclusion criteria; 1568 patients were included in our meta-analysis of significant fibrosis and 2058 in that of advanced fibrosis. The area under the summary curve was 0.81 (95% CI 0.77–0.84) in detecting significant fibrosis and 0.79 (95% CI 0.73–0.82) for advanced fibrosis. Our results support Pro-C3 as an important candidate biomarker for non-invasive assessment of liver fibrosis in NAFLD. Further direct comparisons with currently recommended non-invasive tests will demonstrate whether Pro-C3 panels can outperform these tests, and improve care paths for patients with NAFLD.
Collapse
|
778
|
Oliva ME, Ingaramo P, Vega Joubert MB, Ferreira MDR, D'Alessandro ME. Effects of Salvia hispanica L. (chia) seed on blood coagulation, endothelial dysfunction and liver fibrosis in an experimental model of Metabolic Syndrome. Food Funct 2021; 12:12407-12420. [PMID: 34797360 DOI: 10.1039/d1fo02274a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The aim of this study was to analyze blood coagulation, endothelial dysfunction and liver fibrosis in an experimental model of Metabolic Syndrome (MS) induced by chronic administration of a sucrose-rich diet (SRD) and to evaluate the effects of chia seed as a therapeutic strategy. Male Wistar rats were fed with a reference diet (RD) - 6 months - or a SRD - 3 months. Then, the last group was randomly divided into two subgroups. One subgroup continued receiving the SRD for up to 6 months and the other was fed with a SRD where whole chia seed was incorporated as the source of dietary fat for the next 3 months (SRD + CHIA). Results showed that rats fed a SRD for a long period of time develop dyslipidemia, visceral adiposity, insulin resistance, and a hypercoagulable and hypofibrinolytic basal state. Hepatic VCAM-1 (main adhesion molecules involved in endothelial dysfunction) expression was significantly increased. In addition, the SRD group presented hepatic steatosis, a significant increase in interstitial collagen deposition and hydroxyproline content. Liver TGF-β1 (a key cytokine involved in fibrogenesis) levels increased and a negative correlation with PPARα protein mass levels was found. The administration of chia seed for 3 months reversed dyslipidemia, visceral adiposity and insulin resistance. Platelet count, coagulation parameters and plasma fibrinogen levels were normalized. In the liver tissue, VCAM-1 expression, steatosis, interstitial collagen deposition and the hydroxyproline content decreased. TGF-β1 expression was decreased and this was associated with an increase in the PPARα protein levels. The present study showed new aspects in the progression from liver steatosis to fibrosis in dyslipidemic insulin-resistant rats chronically fed a sucrose-rich diet. Chia seed supplementation could be used as a functional food and a potential dietary strategy to prevent or ameliorate disorders related to atherothrombotic cardiovascular events and NASH.
Collapse
Affiliation(s)
- María Eugenia Oliva
- Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Ciudad Universitaria, cc242 (3000) Santa Fe, Argentina.
| | - Paola Ingaramo
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Cs. Biológicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Santa Fe, Argentina
| | - Michelle Berenice Vega Joubert
- Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Ciudad Universitaria, cc242 (3000) Santa Fe, Argentina.
| | - María Del Rosario Ferreira
- Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Ciudad Universitaria, cc242 (3000) Santa Fe, Argentina.
| | - María Eugenia D'Alessandro
- Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Ciudad Universitaria, cc242 (3000) Santa Fe, Argentina.
| |
Collapse
|
779
|
Xin X, Cheng C, Bei-Yu C, Hong-Shan L, Hua-Jie T, Xin W, Zi-Ming A, Qin-Mei S, Yi-Yang H, Qin F. Caffeine and EGCG Alleviate High-Trans Fatty Acid and High-Carbohydrate Diet-Induced NASH in Mice: Commonality and Specificity. Front Nutr 2021; 8:784354. [PMID: 34881283 PMCID: PMC8647766 DOI: 10.3389/fnut.2021.784354] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 10/27/2021] [Indexed: 12/19/2022] Open
Abstract
Caffeine and epigallocatechin-3-gallate (EGCG), which respectively, are the main functional extracts from coffee and green tea, and present protective effects against non-alcoholic fatty liver diseases (NAFLD). These two beverages and their functional extracts are highly recommended as potential treatments for obesity and NAFLD in clinics; however, their pharmacodynamic effects and pharmacological mechanisms in non-alcoholic steatohepatitis (NASH) remain unclear. Therefore, the aim of this study was to explore the commonality and specificity of the pharmacodynamic effects and pharmacological mechanisms of caffeine and EGCG on NASH mice, which were fed with a high-trans fatty acid/high-carbohydrate (HFHC) diet. C57BL/6J mice were fed a normal diet (control group) or an HFHC diet (HFHC group) for 24 weeks. HFHC group mice were additionally treated with caffeine (75 mg/kg) or EGCG (100 mg/kg) for 6 weeks, using obeticholic acid (OCA,10 mg/kg) as a positive control group. The pharmacological effects of the drugs, including effects on glucose and lipid metabolism and liver inflammation and fibrosis, were evaluated. Gene expression in liver tissue samples from the different groups were assessed. Both caffeine and EGCG significantly reduced the liver manifestations of NASH induced by HFHC. The pathological aspects of liver lipid deposition, inflammation, and liver fibrosis in both groups were strongly ameliorated. Of note, most indexes were strongly reversed in the caffeine group, although AST activity, fasting blood glucose, and the HOMA-IR index were improved in the ECGC group. There were 714 differentially expressed genes between the caffeine and HFHC groups and 268 differentially expressed genes between the EGCG and HFHC groups. Twenty and 17 NASH-related KEGG signaling pathways were enriched by caffeine and EGCG. This study confirmed that 75 mg/kg caffeine and 100 mg/kg EGCG could significantly improve liver lipid deposition, glucose metabolism, inflammation, and fibrosis in a mouse model of NASH induced by HFHC. The bioinformatics platform we built for caffeine and EGCG in NASH disease found that the two drugs may greatly overlap in improving the mechanism related to NASH inflammation. However, caffeine may have better potential in regulating glucose metabolism and EGCG may have better potential in regulating lipid metabolism.
Collapse
Affiliation(s)
- Xin Xin
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chen Cheng
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Cai Bei-Yu
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li Hong-Shan
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tian Hua-Jie
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wang Xin
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - An Zi-Ming
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Sun Qin-Mei
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hu Yi-Yang
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China.,Key Laboratory of Liver and Kidney Diseases, Shanghai University of Traditional Chinese Medicine, Ministry of Education, Shanghai, China
| | - Feng Qin
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China.,Key Laboratory of Liver and Kidney Diseases, Shanghai University of Traditional Chinese Medicine, Ministry of Education, Shanghai, China
| |
Collapse
|
780
|
The New Therapeutic Approaches in the Treatment of Non-Alcoholic Fatty Liver Disease. Int J Mol Sci 2021; 22:ijms222413219. [PMID: 34948020 PMCID: PMC8704688 DOI: 10.3390/ijms222413219] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/25/2021] [Accepted: 12/02/2021] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most prevalent chronic liver disease which is characterized by extremely complex pathogenetic mechanisms and multifactorial etiology. Some of the many pathophysiological mechanisms involved in the development of NAFLD include oxidative stress, impaired mitochondrial metabolism, inflammation, gut microbiota, and interaction between the brain-liver-axis and the regulation of hepatic lipid metabolism. The new therapeutic approaches in the treatment of NAFLD are targeting some of these milestones along the pathophysiological pathway and include drugs like agonists of peroxisome proliferator-activated receptors (PPARs), glucagon-like peptide-1 (GLP-1) agonists, sodium/glucose transport protein 2 (SGLT2) inhibitors, farnesoid X receptor (FXR) agonists, probiotics, and symbiotics. Further efforts in biomedical sciences should focus on the investigation of the relationship between the microbiome, liver metabolism, and response to inflammation, systemic consequences of metabolic syndrome.
Collapse
|
781
|
Huang H, Wang Q, Shi X, Chen Y, Shen C, Zhang J, Xu C. Association between Monocyte to High-Density Lipoprotein Cholesterol Ratio and Nonalcoholic Fatty Liver Disease: A Cross-Sectional Study. Mediators Inflamm 2021; 2021:6642246. [PMID: 34916874 PMCID: PMC8670965 DOI: 10.1155/2021/6642246] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 11/02/2021] [Accepted: 11/03/2021] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND The aim of the present study was to investigate the association between monocyte to high-density lipoprotein cholesterol ratio (MHR) and nonalcoholic fatty liver disease (NAFLD) in Chinese population. METHODS We enrolled 14189 individuals who attended their annual health examinations in the study. We performed the anthropometric and laboratory measurements and diagnosed NAFLD by hepatic ultrasonography without evidence of other etiologies of chronic liver disease. Student's t-test, Mann-Whitney U test, and chi-squared (χ 2) test was used to compare the differences of clinical characteristics between participants with or without NAFLD. Pearson's and Spearman's analyses were performed to assess the correlation of MHR and NAFLD risk factors. Univariate and multivariate logistic regression analyses were conducted to explore whether MHR associated with NAFLD. RESULTS Thirty-five percent of the participants enrolled were diagnosed with NAFLD. Compared with healthy controls, NAFLD patients were male predominant, older, and had higher body mass index, waist circumference, and systolic and diastolic blood pressure, as well as higher levels of alanine aminotransferase, aspartate aminotransferase, γ-glutamyl transferase, triglyceride, total cholesterol, low-density lipoprotein cholesterol, fasting plasma glucose, glycated hemoglobin A1c, and serum uric acid, but lower levels of serum high-density lipoprotein cholesterol. Besides, MHR was significantly higher in NAFLD patients than healthy controls [5.35 (4.18-6.84) versus 4.53 (3.48-5.93), P < 0.001]. MHR quartiles were positively related to the prevalence of NAFLD (P < 0.001 for trend). In multivariate logistic regression analysis, MHR was positively associated with the risk of NAFLD after adjusting age, gender, body mass index, waist circumference, diastolic blood pressure, alanine aminotransferase, triglyceride, total cholesterol, fasting plasma glucose, and serum uric acid (OR: 1.026, 95% CI: 1.002-1.052; P = 0.037). CONCLUSIONS MHR is significantly and positively associated with the risk of NAFLD.
Collapse
Affiliation(s)
- Hangkai Huang
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Qinqiu Wang
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xiaoying Shi
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yishu Chen
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Chao Shen
- Health Management Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Juanwen Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Chengfu Xu
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| |
Collapse
|
782
|
Li H, Wang Q, Chen P, Zhou C, Zhang X, Chen L. Ursodeoxycholic Acid Treatment Restores Gut Microbiota and Alleviates Liver Inflammation in Non-Alcoholic Steatohepatitic Mouse Model. Front Pharmacol 2021; 12:788558. [PMID: 34938193 PMCID: PMC8685972 DOI: 10.3389/fphar.2021.788558] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 11/05/2021] [Indexed: 12/12/2022] Open
Abstract
Gut microbiota dysbiosis plays an important role in the progression of non-alcoholic fatty liver disease (NAFLD), and no approved drugs are available for NAFLD treatment. In this study, we aimed to explore the dynamic changes of gut microbiota at the different stages of NAFLD and determine whether ursodeoxycholic acid (UDCA) could improve liver histopathological features of non-alcoholic steatohepatitis (NASH) mice induced by a high-fat high-cholesterol (HFHC) diet and its impact on gut microbiota. 6-week-old male C57BL/6 mice were fed with a HFHC or normal diet for 12, 18, and 24 weeks, respectively, to simulate the different stages of NAFLD. 16s ribosomal RNA genes from mice fecal samples at the different time points were sequenced to evaluate the dynamic changes of the gut microbiota. Then, C57BL/6 mice were fed with a HFHC diet for 24 weeks to establish the NASH model. Different doses of UDCA were administered intragastrically for additional 4 weeks. Normal diet-fed mice were taken as control. Serum samples, liver, and intestine tissues were harvested for biochemical tests and histopathological examinations. 16s ribosomal RNA genes from mice fecal samples were sequenced to assess the structural changes of gut microbiota. HFHC diet-fed mice developed simple steatosis, steatohepatitis, and fibrosis at 12, 18, and 24 weeks, respectively. The profile of gut microbiota dynamically changed with the different stages of NAFLD. NASH mice had significantly higher abundance of Fecalibaculum, Coriobacteriaceae_UCG-002, and Enterorhabdus, and lower abundance of norank_f_Muribaculaceae, Bacteroides, and Alistipes, which were partially restored by UDCA treatment. UDCA treatment significantly attenuated hepatic inflammation of NASH mice as indicated by the sum of ballooning and lobular inflammation of the NALFD activity score (3.2 ± 0.8 vs 1.8 ± 0.8, p = 0.029), and partially restored gut microbiota dysbiosis, and increased the expression of Claudin-1 and ZO-1 in the intestine, but did not activate the suppressed Farnesoid X receptor signal pathway. Conclusions: The gut microbiota dynamically changes with the different stages of NAFLD. UDCA treatment (120 mg/kg) could partially restore gut microbiota, repair gut barrier integrity, and attenuate hepatic inflammation in the NASH mouse model.
Collapse
Affiliation(s)
- Hu Li
- Department of Gastroenterology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Infectious Disease, Shanghai Jiao Tong University Affiliated Sixth People`s Hospital, Shanghai, China
| | - Qingling Wang
- Department of Infectious Disease, Research Laboratory of Clinical Virology, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
- Department of Infectious Disease, The First People’s Hospital of Yunnan Province, Kunming, China
| | - Peizhan Chen
- Central Laboratory, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chenghua Zhou
- Central Laboratory, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xinxin Zhang
- Department of Infectious Disease, Research Laboratory of Clinical Virology, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Li Chen
- Department of Gastroenterology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
783
|
Lai JL, Lian YE, Wu JY, Wang YD, Bai YN. Verapamil induces autophagy to improve liver regeneration in non-alcoholic fatty liver mice. Adipocyte 2021; 10:532-545. [PMID: 34699301 PMCID: PMC8555529 DOI: 10.1080/21623945.2021.1983241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 12/13/2022] Open
Abstract
Verapamil can restore intracellular calcium homeostasis, increase the fusion of autophagosomes and lysosomes, reduce lipid droplet accumulation and inhibit inflammation and insulin resistance in high-fat-fed mice. The present study aimed to investigate verapamil's effect and its underlying liver regeneration mechanism in mice with non-alcoholic fatty liver. After 50% hepatectomy was performed, the changes of autophagy and liver regeneration were evaluated by detecting cell proliferation and autophagy at each time point. Then, 25mg/kg verapamil was injected intraperitoneally for 10 d before an operation in the mild to moderate fatty liver and severe fatty liver groups. The control group and mild to moderate fatty liver group reached the peak of proliferation at 24-48h after operation, and the mice with severe fatty liver and steatohepatitis reached the peak at 48-72h. Autophagy in the normal group and mild to moderate fatty liver group reached the peak 48 hours after operation. Verapamil injection can enhance autophagy, reduce the weight of fatty liver mice, improve liver function and liver regeneration. Verapamil can induce autophagy, improve hepatocyte function and promote hepatocyte regeneration through the mTOR independent signaling pathway, thus improving the process of liver regeneration after partial hepatectomy.
Collapse
Affiliation(s)
- Jian-Lin Lai
- Shengli Clinical Medical College of Fujian Medical University, Department of Hepatobiliary and Pancreatic Surgery, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Yuan-E Lian
- Department of Pathology, The Affiliated Union Hospital of Fujian Medical University, Fuzhou, 350001, China
| | - Jun-Yi Wu
- Shengli Clinical Medical College of Fujian Medical University, Department of Hepatobiliary and Pancreatic Surgery, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Yao-Dong Wang
- Shengli Clinical Medical College of Fujian Medical University, Department of Hepatobiliary and Pancreatic Surgery, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Yan-Nan Bai
- Shengli Clinical Medical College of Fujian Medical University, Department of Hepatobiliary and Pancreatic Surgery, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| |
Collapse
|
784
|
Tilg H, Adolph TE, Dudek M, Knolle P. Non-alcoholic fatty liver disease: the interplay between metabolism, microbes and immunity. Nat Metab 2021; 3:1596-1607. [PMID: 34931080 DOI: 10.1038/s42255-021-00501-9] [Citation(s) in RCA: 257] [Impact Index Per Article: 64.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 11/04/2021] [Indexed: 02/07/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has emerged pandemically across the globe and particularly affects patients with obesity and type 2 diabetes. NAFLD is a complex systemic disease that is characterised by hepatic lipid accumulation, lipotoxicity, insulin resistance, gut dysbiosis and inflammation. In this review, we discuss how metabolic dysregulation, the gut microbiome, innate and adaptive immunity and their interplay contribute to NAFLD pathology. Lipotoxicity has been shown to instigate liver injury, inflammation and insulin resistance. Synchronous metabolic dysfunction, obesity and related nutritional perturbation may alter the gut microbiome, in turn fuelling hepatic and systemic inflammation by direct activation of innate and adaptive immune responses. We review evidence suggesting that, collectively, these unresolved exogenous and endogenous cues drive liver injury, culminating in liver fibrosis and advanced sequelae of this disorder such as liver cirrhosis and hepatocellular carcinoma. Understanding NAFLD as a complex interplay between metabolism, gut microbiota and the immune response will challenge the clinical perception of NAFLD and open new therapeutic avenues.
Collapse
Affiliation(s)
- Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| | - Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Michael Dudek
- Institute of Molecular Immunology and Experimental Oncology, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Percy Knolle
- Institute of Molecular Immunology and Experimental Oncology, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| |
Collapse
|
785
|
Córdova-Gallardo J, Keaveny AP, Qi X, Méndez-Sánchez N. Metabolic associated fatty liver disease and acute-on-chronic liver failure: common themes for common problems. Eur J Gastroenterol Hepatol 2021; 33:e84-e93. [PMID: 34985050 DOI: 10.1097/meg.0000000000002335] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Metabolic associated fatty liver disease (MAFLD) affects 20-30% of the worldwide population and is becoming the most common cause of chronic liver disease, cirrhosis and hepatocellular carcinoma (HCC). MAFLD is the hepatic expression of metabolic dysfunction correlated with a variety of metabolic comorbidities including obesity, dyslipidemia, hypertension and type 2 diabetes (T2DM). Obesity, altered gut permeability, chronic inflammation and dysbiosis related to MAFLD might predispose patients with cirrhosis to the development of acute-on-chronic liver failure (ACLF); however, this relationship remains unclear. ACLF is a syndrome with high short-term mortality, presenting with acute hepatic decompensation associated with organ failures in patients with underlying chronic liver disease with or without an identifiable precipitating event. While this syndrome can occur in any patient with cirrhosis, the increasing prevalence of cirrhosis due to MAFLD is of great concern because, in a recent analysis, MAFLD was the fastest rising cause of cirrhosis associated with ACLF among patients listed for LT in the US. In this review, we will discuss the current knowledge on MAFLD and the development of ACLF.
Collapse
Affiliation(s)
- Jacqueline Córdova-Gallardo
- Department of Hepatology, Service of Surgery and Obesity Clinic, General Hospital "Dr. Manuel Gea González"
- Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - Andrew P Keaveny
- Department of Transplantation, Mayo Clinic, Jacksonville, Florida, USA
| | - Xingshun Qi
- General Hospital of Northern Theater Command
- Shenyang Pharmaceutical University, Shenyang, China
| | - Nahum Méndez-Sánchez
- Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
- Liver Research Unit, Medica Sur Clinic & Foundation, Mexico City, Mexico
| |
Collapse
|
786
|
Rios RS, Zheng KI, Zheng MH. Non-alcoholic steatohepatitis and risk of hepatocellular carcinoma. Chin Med J (Engl) 2021; 134:2911-2921. [PMID: 34855640 PMCID: PMC8710331 DOI: 10.1097/cm9.0000000000001888] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Indexed: 12/18/2022] Open
Abstract
ABSTRACT The emergence of non-alcoholic fatty liver disease (NAFLD) as the leading chronic liver disease worldwide raises some concerns. In particular, NAFLD is closely tied to sedentary lifestyle habits and associated with other metabolic diseases, such as obesity and diabetes. At the end of the disease spectrum, non-alcoholic steatohepatitis (NASH) may progress to cirrhosis and hepatocellular carcinoma (HCC), representing a serious health problem to modern society. Recently, an increasing number of HCC cases originating from this progressive disease spectrum have been identified, with different levels of severity and complications. Updating the current guidelines by placing a bigger focus on this emerging cause and highlighting some of its unique features is necessary. Since, the drivers of the disease are complex and multifactorial, in order to improve future outcomes, having a better understanding of NASH progression into HCC may be helpful. The risks that can promote disease progression and currently available management strategies employed to monitor and treat NASH-related HCC make up the bulk of this review.
Collapse
Affiliation(s)
- Rafael S. Rios
- NAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Kenneth I. Zheng
- NAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Ming-Hua Zheng
- NAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Institute of Hepatology, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Key Laboratory of Diagnosis and Treatment for The Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, Zhejiang 325000, China
| |
Collapse
|
787
|
NASH y cirugía metabólica: cuando tienes los datos pero no puedes demostrarlo. Cir Esp 2021. [DOI: 10.1016/j.ciresp.2021.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
788
|
Dark and bright side of targeting fibroblast growth factor receptor 4 in the liver. J Hepatol 2021; 75:1440-1451. [PMID: 34364916 DOI: 10.1016/j.jhep.2021.07.029] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/09/2021] [Accepted: 07/26/2021] [Indexed: 12/12/2022]
Abstract
Fibroblast growth factor (FGF) receptor 4 (FGFR4) and its cognate ligand, FGF19, are implicated in a range of cellular processes, including differentiation, metabolism and proliferation. Indeed, their aberrant activation has been associated with the development of hepatic tumours. Despite great advances in early diagnosis and the development of new therapies, liver cancer is still associated with a high mortality rate, owing primarily to high molecular heterogeneity and unclear molecular targeting. The development of FGFR4 inhibitors is a promising tool in patients with concomitant supraphysiological levels of FGF19 and several clinical trials are testing these treatments for patients with advanced hepatocellular carcinoma (HCC). Conversely, using FGF19 analogues to activate FGFR4-KLOTHO β represents a novel therapeutic strategy in patients presenting with cholestatic liver disorders and non-alcoholic steatohepatitis, which could potentially prevent the development of metabolic HCC. Herein, we provide an overview of the currently available therapeutic options for targeting FGFR4 in HCC and other liver diseases, highlighting the need to carefully stratify patients and personalise therapeutic strategies.
Collapse
|
789
|
Sekhon AK, Gollapalli A, Kaur D, Janssen B, Stevens ML, Valerio F, Sierra-Hoffman MA. A New Potential Strategy for Acute Non-Alcoholic Steatohepatitis (NASH). AMERICAN JOURNAL OF CASE REPORTS 2021; 22:e932961. [PMID: 34826302 PMCID: PMC8635219 DOI: 10.12659/ajcr.932961] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 10/23/2021] [Accepted: 09/20/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver disease in the United States, and 25% of patients with NAFLD progress to non-alcoholic steatohepatitis (NASH). NAFLD is predicted to be the most common indication for liver transplantation by 2030. Despite associated high morbidity and mortality, there is currently no approved therapy for NASH. PCSK9 inhibitors are approved for reducing LDL in patients who are statin-intolerant or need further LDL reduction. Increased LDL levels are independently associated with an elevated risk of NAFLD. CASE REPORT We present a case of a 39-year-old woman with acute NASH with familial hypercholesterolemia that was refractory to lifestyle modifications and HMG-CoA reductase inhibitors. An episode of rhabdomyolysis warranted a search for alternatives to statin therapy. Results of a liver biopsy showed microvesicular and macrovesicular steatosis with ballooning degeneration, indicating acute NASH. She was started on PCSK9 inhibitors as salvage therapy. Three monthly doses resulted in a more than an 80% reduction in ALT and AST and a 48% reduction in LDL levels. A liver biopsy done 8 months after the first biopsy showed normalization of liver histology. CONCLUSIONS The use of PCSK9 inhibitors showed a dramatic response in this patient who failed conventional therapies, and the encouraging results seen in this case merit further research into the use of PCSK9 inhibitors as first-line therapy for the acute phase of NASH.
Collapse
Affiliation(s)
| | | | - Dharamjeet Kaur
- Research Department, Texas A&M-affiliated DeTar Family Medicine Residency Program, Victoria, TX, USA
| | - Bryan Janssen
- Department of Pathology, DeTar Hospital, Victoria, TX, USA
| | - Mark L. Stevens
- Research Department, Texas A&M-affiliated DeTar Family Medicine Residency Program, Victoria, TX, USA
| | - Fernando Valerio
- Department of Critical Care, Hospital CEMESA, San Pedro Sula, Cortés, Honduras
| | - Miguel A. Sierra-Hoffman
- Research Department, Texas A&M-affiliated DeTar Family Medicine Residency Program, Victoria, TX, USA
| |
Collapse
|
790
|
Aminian A, Al-Kurd A, Wilson R, Bena J, Fayazzadeh H, Singh T, Albaugh VL, Shariff FU, Rodriguez NA, Jin J, Brethauer SA, Dasarathy S, Alkhouri N, Schauer PR, McCullough AJ, Nissen SE. Association of Bariatric Surgery With Major Adverse Liver and Cardiovascular Outcomes in Patients With Biopsy-Proven Nonalcoholic Steatohepatitis. JAMA 2021; 326:2031-2042. [PMID: 34762106 PMCID: PMC8587225 DOI: 10.1001/jama.2021.19569] [Citation(s) in RCA: 192] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
IMPORTANCE No therapy has been shown to reduce the risk of serious adverse outcomes in patients with nonalcoholic steatohepatitis (NASH). OBJECTIVE To investigate the long-term relationship between bariatric surgery and incident major adverse liver outcomes and major adverse cardiovascular events (MACE) in patients with obesity and biopsy-proven fibrotic NASH without cirrhosis. DESIGN, SETTING, AND PARTICIPANTS In the SPLENDOR (Surgical Procedures and Long-term Effectiveness in NASH Disease and Obesity Risk) study, of 25 828 liver biopsies performed at a US health system between 2004 and 2016, 1158 adult patients with obesity were identified who fulfilled enrollment criteria, including confirmed histological diagnosis of NASH and presence of liver fibrosis (histological stages 1-3). Baseline clinical characteristics, histological disease activity, and fibrosis stage of patients who underwent simultaneous liver biopsy at the time of bariatric surgery were balanced with a nonsurgical control group using overlap weighting methods. Follow-up ended in March 2021. EXPOSURES Bariatric surgery (Roux-en-Y gastric bypass, sleeve gastrectomy) vs nonsurgical care. MAIN OUTCOMES AND MEASURES The primary outcomes were the incidence of major adverse liver outcomes (progression to clinical or histological cirrhosis, development of hepatocellular carcinoma, liver transplantation, or liver-related mortality) and MACE (a composite of coronary artery events, cerebrovascular events, heart failure, or cardiovascular death), estimated using the Firth penalized method in a multivariable-adjusted Cox regression analysis framework. RESULTS A total of 1158 patients (740 [63.9%] women; median age, 49.8 years [IQR, 40.9-57.9 years], median body mass index, 44.1 [IQR, 39.4-51.4]), including 650 patients who underwent bariatric surgery and 508 patients in the nonsurgical control group, with a median follow-up of 7 years (IQR, 4-10 years) were analyzed. Distribution of baseline covariates, including histological severity of liver injury, was well-balanced after overlap weighting. At the end of the study period in the unweighted data set, 5 patients in the bariatric surgery group and 40 patients in the nonsurgical control group experienced major adverse liver outcomes, and 39 patients in the bariatric surgery group and 60 patients in the nonsurgical group experienced MACE. Among the patients analyzed with overlap weighting methods, the cumulative incidence of major adverse liver outcomes at 10 years was 2.3% (95% CI, 0%-4.6%) in the bariatric surgery group and 9.6% (95% CI, 6.1%-12.9%) in the nonsurgical group (adjusted absolute risk difference, 12.4% [95% CI, 5.7%-19.7%]; adjusted hazard ratio, 0.12 [95% CI, 0.02-0.63]; P = .01). The cumulative incidence of MACE at 10 years was 8.5% (95% CI, 5.5%-11.4%) in the bariatric surgery group and 15.7% (95% CI, 11.3%-19.8%) in the nonsurgical group (adjusted absolute risk difference, 13.9% [95% CI, 5.9%-21.9%]; adjusted hazard ratio, 0.30 [95% CI, 0.12-0.72]; P = .007). Within the first year after bariatric surgery, 4 patients (0.6%) died from surgical complications, including gastrointestinal leak (n = 2) and respiratory failure (n = 2). CONCLUSIONS AND RELEVANCE Among patients with NASH and obesity, bariatric surgery, compared with nonsurgical management, was associated with a significantly lower risk of incident major adverse liver outcomes and MACE.
Collapse
Affiliation(s)
- Ali Aminian
- Bariatric and Metabolic Institute, Department of General Surgery, Cleveland Clinic, Cleveland, Ohio
| | - Abbas Al-Kurd
- Bariatric and Metabolic Institute, Department of General Surgery, Cleveland Clinic, Cleveland, Ohio
| | - Rickesha Wilson
- Bariatric and Metabolic Institute, Department of General Surgery, Cleveland Clinic, Cleveland, Ohio
| | - James Bena
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Hana Fayazzadeh
- Bariatric and Metabolic Institute, Department of General Surgery, Cleveland Clinic, Cleveland, Ohio
| | - Tavankit Singh
- Division of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, Ohio
- Department of Gastroenterology and Hepatology, Allegheny General Hospital, Pittsburgh, Pennsylvania
| | - Vance L. Albaugh
- Bariatric and Metabolic Institute, Department of General Surgery, Cleveland Clinic, Cleveland, Ohio
- Bariatric and Metabolic Institute, Pennington Biomedical Research Center, Baton Rouge, Louisiana
| | - Faiz U. Shariff
- Bariatric and Metabolic Institute, Department of General Surgery, Cleveland Clinic, Cleveland, Ohio
- Department of Surgery, Wellspan Hospital, York, Pennsylvania
| | - Noe A. Rodriguez
- Bariatric and Metabolic Institute, Department of General Surgery, Cleveland Clinic, Cleveland, Ohio
- Department of Surgery, Northwest Medical Center, Springdale, Arkansas
| | - Jian Jin
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Stacy A. Brethauer
- Bariatric and Metabolic Institute, Department of General Surgery, Cleveland Clinic, Cleveland, Ohio
- Department of Surgery, Wexner Medical Center, Ohio State University, Columbus
| | | | - Naim Alkhouri
- Division of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, Ohio
- Fatty Liver Program, Arizona Liver Health, Chandler
| | - Philip R. Schauer
- Bariatric and Metabolic Institute, Department of General Surgery, Cleveland Clinic, Cleveland, Ohio
- Bariatric and Metabolic Institute, Pennington Biomedical Research Center, Baton Rouge, Louisiana
| | | | - Steven E. Nissen
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
791
|
Mazzini GS, Augustin T, Noria S, Romero-Marrero C, Li N, Hameed B, Eisenberg D, Azagury DE, Ikramuddin S. ASMBS Position Statement on the Impact of Metabolic and Bariatric Surgery on Nonalcoholic Steatohepatitis. Surg Obes Relat Dis 2021; 18:314-325. [PMID: 34953742 DOI: 10.1016/j.soard.2021.11.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 11/07/2021] [Indexed: 10/25/2022]
Affiliation(s)
- Guilherme S Mazzini
- Division of Bariatric and Gastrointestinal Surgery, Department of Surgery, Virginia Commonwealth University, Richmond, Virginia
| | - Toms Augustin
- Department of General Surgery, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, Ohio
| | - Sabrena Noria
- Department of Surgery, Division of General and Gastrointestinal Surgery, The Ohio state University, Wexner Medical Center, Columbus, Ohio
| | - Carlos Romero-Marrero
- South Florida Transplant Center, Broward Health Medical Center, Fort Lauderdale, Florida
| | - Na Li
- Division of Gastroenterology, Hepatology, & Nutrition, Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Bilal Hameed
- Department of Medicine, University of California, San Francisco, California
| | - Dan Eisenberg
- Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Dan E Azagury
- Section of Minimally Invasive & Bariatric Surgery, Stanford University School of Medicine, Stanford, California
| | - Sayeed Ikramuddin
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
792
|
Perez-Carreras M, Casis-Herce B, Rivera R, Fernandez I, Martinez-Montiel P, Villena V. Non-alcoholic fatty liver disease in patients with intestinal, pulmonary or skin diseases: Inflammatory cross-talk that needs a multidisciplinary approach. World J Gastroenterol 2021; 27:7113-7124. [PMID: 34887631 PMCID: PMC8613653 DOI: 10.3748/wjg.v27.i41.7113] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/04/2021] [Accepted: 09/16/2021] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is currently considered the most common cause of liver disease. Its prevalence is increasing in parallel with the obesity and type 2 diabetes mellitus (DM2) epidemics in developed countries. Several recent studies have suggested that NAFLD may be the hepatic manifestation of a systemic inflammatory metabolic disease that also affects other organs, such as intestine, lungs, skin and vascular endothelium. It appears that local and systemic proinflammatory/anti-inflammatory cytokine imbalance, together with insulin resistance and changes in the intestinal microbiota, are pathogenic mechanisms shared by NAFLD and other comorbidities. NAFLD is more common in patients with extrahepatic diseases such as inflammatory bowel disease (IBD), obstructive syndrome apnea (OSA) and psoriasis than in the general population. Furthermore, there is evidence that this association has a negative impact on the severity of liver lesions. Specific risk characteristics for NAFLD have been identified in populations with IBD (i.e. age, obesity, DM2, previous bowel surgery, IBD evolution time, methotrexate treatment), OSA (i.e. obesity, DM2, OSA severity, increased transaminases) and psoriasis (i.e. age, metabolic factors, severe psoriasis, arthropathy, elevated transaminases, methotrexate treatment). These specific phenotypes might be used by gastroenterologists, pneumologists and dermatologists to create screening algorithms for NAFLD. Such algorithms should include non-invasive markers of fibrosis used in NAFLD to select subjects for referral to the hepatologist. Prospective, controlled studies in NAFLD patients with extrahepatic comorbidities are required to demonstrate a causal relationship and also that appropriate multidisciplinary management improves these patients’ prognosis and survival.
Collapse
Affiliation(s)
- Mercedes Perez-Carreras
- Gastroenterology and Hepatology Unit, 12 de Octubre Universitary Hospital, Madrid 28041, Spain
- Faculty of Medicine, Complutense University, Madrid 28040, Spain
| | - Begoña Casis-Herce
- Gastroenterology and Hepatology Unit, 12 de Octubre Universitary Hospital, Madrid 28041, Spain
- Faculty of Medicine, Complutense University, Madrid 28040, Spain
| | - Raquel Rivera
- Faculty of Medicine, Complutense University, Madrid 28040, Spain
- Dermatology Department, 12 de Octubre Universitary Hospital, Madrid 28041, Spain
| | - Inmaculada Fernandez
- Gastroenterology and Hepatology Unit, 12 de Octubre Universitary Hospital, Madrid 28041, Spain
- Faculty of Medicine, Complutense University, Madrid 28040, Spain
| | - Pilar Martinez-Montiel
- Gastroenterology and Hepatology Unit, 12 de Octubre Universitary Hospital, Madrid 28041, Spain
- Faculty of Medicine, Complutense University, Madrid 28040, Spain
| | - Victoria Villena
- Faculty of Medicine, Complutense University, Madrid 28040, Spain
- Pneumology Service, 12 de Octubre Universitary Hospital, Madrid 28041, Spain
| |
Collapse
|
793
|
Fengler VH, Macheiner T, Goessler W, Ratzer M, Haybaeck J, Sargsyan K. Hepatic Response of Magnesium-Restricted Wild Type Mice. Metabolites 2021; 11:metabo11110762. [PMID: 34822420 PMCID: PMC8625093 DOI: 10.3390/metabo11110762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/26/2021] [Accepted: 11/03/2021] [Indexed: 11/16/2022] Open
Abstract
Magnesium-deficiency is implicated in many metabolic disorders, e.g., type 2 diabetes and metabolic syndrome, representing risk factors for non-alcoholic fatty liver disease (NAFLD). This study aims to investigate the contribution of magnesium-restriction to the development of NAFLD. Magnesium-deficiency was induced in C57BL/6 mice by feeding a magnesium-deficient-diet. Metabolic markers as well as markers of inflammation and liver function were assessed. Furthermore, liver tissue was examined histopathologically and compared with specimens from high-fat-diet fed and control mice. Finally, the hepatic inflammatory response was quantified by determining hepatic IL-6, TNFα, and MCP-1. Magnesium-restriction resulted in at least a 2-fold significant reduction of serum magnesium levels compared to the high-fat-diet fed and control mice, whereas the hepatic magnesium content was decreased due to high-fat-diet feeding. No changes in metabolic markers in magnesium-restricted mice were observed, while the cholesterol content was elevated in high-fat-diet fed mice. Magnesium-restricted mice additionally featured inflammation and enlarged hepatocytes in liver histology. Furthermore, magnesium-restricted and high-fat-diet fed mice exhibited elevated hepatic TNFα levels compared to control mice. Accordingly, our data suggest that magnesium is involved in hepatic inflammatory processes and hepatocyte enlargement, key histological features of human NAFLD, and may therefore contribute to development and progression of the disease.
Collapse
Affiliation(s)
- Vera H. Fengler
- Biobank Graz, Medical University of Graz, 8036 Graz, Austria;
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Tanja Macheiner
- International Biobank and Education, Medical University of Graz, 8036 Graz, Austria;
| | - Walter Goessler
- Institute of Chemistry, University of Graz, 8010 Graz, Austria;
| | - Maria Ratzer
- Institute for Biomedicine and Health Sciences, Joanneum Research, 8010 Graz, Austria;
| | - Johannes Haybaeck
- Department of Pathology, Neuropathology and Molecular Pathology, Medical University of Innsbruck, 6020 Innsbruck, Austria;
- Diagnostic & Research Center for Molecular BioMedicine, Institute of Pathology, Medical University of Graz, 8036 Graz, Austria
| | - Karine Sargsyan
- International Biobank and Education, Medical University of Graz, 8036 Graz, Austria;
- Correspondence: ; Tel.: +43-316-385-72718
| |
Collapse
|
794
|
Loomba R, Mohseni R, Lucas KJ, Gutierrez JA, Perry RG, Trotter JF, Rahimi RS, Harrison SA, Ajmera V, Wayne JD, O'Farrell M, McCulloch W, Grimmer K, Rinella M, Wai-Sun Wong V, Ratziu V, Gores GJ, Neuschwander-Tetri BA, Kemble G. TVB-2640 (FASN Inhibitor) for the Treatment of Nonalcoholic Steatohepatitis: FASCINATE-1, a Randomized, Placebo-Controlled Phase 2a Trial. Gastroenterology 2021; 161:1475-1486. [PMID: 34310978 DOI: 10.1053/j.gastro.2021.07.025] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/17/2021] [Accepted: 07/13/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND & AIMS Increased de novo lipogenesis creates excess intrahepatic fat and lipotoxins, propagating liver damage in nonalcoholic steatohepatitis. TVB-2640, a fatty acid synthase inhibitor, was designed to reduce excess liver fat and directly inhibit inflammatory and fibrogenic pathways. We assessed the safety and efficacy of TVB-2640 in patients with nonalcoholic steatohepatitis in the United States. METHODS 3V2640-CLIN-005 (FASCINATE-1) was a randomized, placebo-controlled, single-blind study at 10 US sites. Adults with ≥8% liver fat, assessed by magnetic resonance imaging proton density fat fraction, and evidence of liver fibrosis by magnetic resonance elastography ≥2.5 kPa or liver biopsy were eligible. Ninety-nine patients were randomized to receive placebo or 25 mg or 50 mg of TVB-2640 (orally, once-daily for 12 weeks). The primary end points of this study were safety and relative change in liver fat after treatment. RESULTS Liver fat increased in the placebo cohort by 4.5% relative to baseline; in contrast TVB-2640 reduced liver fat by 9.6% in the 25-mg cohort (n = 30; least squares mean: -15.5%; 95% confidence interval, -31.3 to -0.23; P = .053), and 28.1% in the 50-mg cohort (n = 28; least squares mean: -28.0%; 95% confidence interval, -44.5 to -11.6; P = .001). Eleven percent of patients in the placebo group achieved a ≥30% relative reduction of liver fat compared to 23% in the 25-mg group, and 61% in the 50-mg group (P < .001). Secondary analyses showed improvements of metabolic, pro-inflammatory and fibrotic markers. TVB-2640 was well tolerated; adverse events were mostly mild and balanced among the groups. CONCLUSIONS TVB-2640 significantly reduced liver fat and improved biochemical, inflammatory, and fibrotic biomarkers after 12 weeks, in a dose-dependent manner in patients with nonalcoholic steatohepatitis. ClinicalTrials.gov, Number NCT03938246.
Collapse
Affiliation(s)
- Rohit Loomba
- Nonalcoholic Fatty Liver Disease Research Center, Division of Gastroenterology and Hepatology, Department of Medicine, University of California at San Diego, La Jolla, California.
| | | | | | | | | | - James F Trotter
- Baylor University Medical Center, Texas Digestive Disease Consultants, Dallas, Texas
| | - Robert S Rahimi
- Baylor University Medical Center, Texas Digestive Disease Consultants, Dallas, Texas
| | | | - Veeral Ajmera
- Nonalcoholic Fatty Liver Disease Research Center, Division of Gastroenterology and Hepatology, Department of Medicine, University of California at San Diego, La Jolla, California
| | | | | | | | | | - Mary Rinella
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Vincent Wai-Sun Wong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong
| | - Vlad Ratziu
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Hôpital Pitié-Salpêtriére, Institute for Cardiometabolism and Nutrition, Paris, France
| | - Gregory J Gores
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Brent A Neuschwander-Tetri
- Division of Gastroenterology and Hepatology, Saint Louis University School of Medicine, St Louis, Missouri
| | | |
Collapse
|
795
|
Li X, Ge J, Li Y, Cai Y, Zheng Q, Huang N, Gu Y, Han Q, Li Y, Sun R, Liu R. Integrative lipidomic and transcriptomic study unravels the therapeutic effects of saikosaponins A and D on non-alcoholic fatty liver disease. Acta Pharm Sin B 2021; 11:3527-3541. [PMID: 34900534 PMCID: PMC8642447 DOI: 10.1016/j.apsb.2021.03.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/03/2021] [Accepted: 02/09/2021] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) has become one of the most prominent causes of chronic liver diseases and malignancies. However, few therapy has been approved. Radix Bupleuri (RB) is the most frequently used herbal medicine for the treatment of liver diseases. In the current study, we aim to systemically evaluate the therapeutic effects of saikosaponin A (SSa) and saikosaponin D (SSd), the major bioactive monomers in RB, against NAFLD and to investigate the underlying mechanisms. Our results demonstrated that both SSa and SSd improved diet-induced NAFLD. Integrative lipidomic and transcriptomic analysis revealed that SSa and SSd modulated glycerolipid metabolism by regulating related genes, like Lipe and Lipg. SSd profoundly suppressed the fatty acid biosynthesis by downregulating Fasn and Acaca expression and promoted fatty acid degradation by inducing Acox1 and Cpt1a expression. Bioinformatic analysis further predicted the implication of master transcription factors, including peroxisome proliferator-activated receptor alpha (PPARα), in the protective effects of SSa and SSd. These results were further confirmed in vitro in mouse primary hepatocytes. In summary, our study uncoded the complicated mechanisms underlying the promising anti-steatosis activities of saikosaponins (SSs), and provided critical evidence inspiring the discovery of innovative therapies based on SSa and SSd for the treatment of NAFLD and related complications.
Collapse
|
796
|
Lefebvre P, Staels B. Hepatic sexual dimorphism - implications for non-alcoholic fatty liver disease. Nat Rev Endocrinol 2021; 17:662-670. [PMID: 34417588 DOI: 10.1038/s41574-021-00538-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/06/2021] [Indexed: 12/14/2022]
Abstract
The liver is often thought of as a single functional unit, but both its structural and functional architecture make it highly multivalent and adaptable. In any given physiological situation, the liver can maintain metabolic homeostasis, conduct appropriate inflammatory responses, carry out endobiotic and xenobiotic transformation and synthesis reactions, as well as store and release multiple bioactive molecules. Moreover, the liver is a very resilient organ. This resilience means that chronic liver diseases can go unnoticed for decades, yet culminate in life-threatening clinical complications once the adaptive capacity of the liver is overwhelmed. Non-alcoholic fatty liver disease (NAFLD) predisposes individuals to cirrhosis and increases liver-related and cardiovascular disease-related mortality. This Review discusses the accumulating evidence of sexual dimorphism in NAFLD, which is currently rarely considered in preclinical and clinical studies. Increased awareness of the mechanistic causes of hepatic sexual dimorphism could lead to improved understanding of the biological processes that are dysregulated in NAFLD, to the identification of relevant therapeutic targets and to improved risk stratification of patients with NAFLD undergoing therapeutic intervention.
Collapse
Affiliation(s)
- Philippe Lefebvre
- Université Lille, INSERM, CHU Lille, Institut Pasteur de Lille, Lille, France.
| | - Bart Staels
- Université Lille, INSERM, CHU Lille, Institut Pasteur de Lille, Lille, France
| |
Collapse
|
797
|
Gutiérrez-Cuevas J, Santos A, Armendariz-Borunda J. Pathophysiological Molecular Mechanisms of Obesity: A Link between MAFLD and NASH with Cardiovascular Diseases. Int J Mol Sci 2021; 22:11629. [PMID: 34769060 PMCID: PMC8583943 DOI: 10.3390/ijms222111629] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 09/16/2021] [Indexed: 02/06/2023] Open
Abstract
Obesity is now a worldwide epidemic ensuing an increase in comorbidities' prevalence, such as insulin resistance, type 2 diabetes (T2D), metabolic dysfunction-associated fatty liver disease (MAFLD), nonalcoholic steatohepatitis (NASH), hypertension, cardiovascular disease (CVD), autoimmune diseases, and some cancers, CVD being one of the main causes of death in the world. Several studies provide evidence for an association between MAFLD and atherosclerosis and cardio-metabolic disorders, including CVDs such as coronary heart disease and stroke. Therefore, the combination of MAFLD/NASH is associated with vascular risk and CVD progression, but the underlying mechanisms linking MAFLD/NASH and CVD are still under investigation. Several underlying mechanisms may probably be involved, including hepatic/systemic insulin resistance, atherogenic dyslipidemia, hypertension, as well as pro-atherogenic, pro-coagulant, and pro-inflammatory mediators released from the steatotic/inflamed liver. MAFLD is strongly associated with insulin resistance, which is involved in its pathogenesis and progression to NASH. Insulin resistance is a major cardiovascular risk factor in subjects without diabetes. However, T2D has been considered the most common link between MAFLD/NASH and CVD. This review summarizes the evidence linking obesity with MAFLD, NASH, and CVD, considering the pathophysiological molecular mechanisms involved in these diseases. We also discuss the association of MAFLD and NASH with the development and progression of CVD, including structural and functional cardiac alterations, and pharmacological strategies to treat MAFLD/NASH and cardiovascular prevention.
Collapse
Affiliation(s)
- Jorge Gutiérrez-Cuevas
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, University of Guadalajara, CUCS, Guadalajara 44340, Jalisco, Mexico
| | - Arturo Santos
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Campus Guadalajara, Zapopan 45201, Jalisco, Mexico;
| | - Juan Armendariz-Borunda
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, University of Guadalajara, CUCS, Guadalajara 44340, Jalisco, Mexico
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Campus Guadalajara, Zapopan 45201, Jalisco, Mexico;
| |
Collapse
|
798
|
Gao X, Ruan Y, Zhu X, Lin X, Xin Y, Li X, Mai M, Guo H. Deoxycholic Acid Promotes Pyroptosis in Free Fatty Acid-Induced Steatotic Hepatocytes by Inhibiting PINK1-Mediated Mitophagy. Inflammation 2021; 45:639-650. [PMID: 34674097 DOI: 10.1007/s10753-021-01573-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/28/2021] [Accepted: 09/29/2021] [Indexed: 02/06/2023]
Abstract
Nonalcoholic steatohepatitis (NASH) is the inflammatory subtype of nonalcoholic fatty liver disease (NAFLD), which can lead to liver fibrosis and cirrhosis. Bile acid levels are correlated with markers of hepatic injury in NASH, suggesting a possible role for bile acids in the progression of NAFLD. Here, we examined the role of deoxycholic acid (DCA) in driving steatotic hepatocytes to pyroptosis, a pro-inflammatory form of programmed cell death. HepG2 cells were stimulated with odium oleate and sodium palmitate for modeling steatotic hepatocytes and then treated with DCA alone or in combination with a specific mitophagy agonist, carbonyl cyanide 3-chlorophenylhydrazone (CCCP). Our results showed that DCA dose-dependently induced a pro-inflammatory response in steatotic hepatocytes but had no significant effect on lipid accumulation. Moreover, activation of the NLRP3 inflammasome and pyroptosis were triggered by DCA. Expression levels of the mitophagy markers PTEN-induced kinase 1 (PINK1) and E3 ubiquitin ligase Parkin were significantly diminished by DCA, whereas induction of mitophagy by CCCP prevented DCA-induced inflammatory response and restored the pyroptosis. Collectively, our data showed that DCA-induced pyroptosis involves the inhibition of PINK1-mediated mitophagy and the activation of the NLRP3 inflammasome. These findings provide insight into the association of DCA with mitophagy, pyroptosis, and inflammation in NASH.
Collapse
Affiliation(s)
- Xuebin Gao
- Department of Nutrition, School of Public Health, Guangdong Medical University, Dongguan, 523808, China
| | - Yongdui Ruan
- Department of Traditional Chinese Medicine, the First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523710, China
| | - Xuan Zhu
- Department of Nutrition, School of Public Health, Guangdong Medical University, Dongguan, 523808, China
| | - Xiaozhuan Lin
- Department of Nutrition, School of Public Health, Guangdong Medical University, Dongguan, 523808, China
| | - Yan Xin
- Department of Nutrition, School of Public Health, Guangdong Medical University, Dongguan, 523808, China
| | - Xiang Li
- Department of Nutrition, School of Public Health, Guangdong Medical University, Dongguan, 523808, China
| | - Meiqing Mai
- Department of Nutrition, School of Public Health, Guangdong Medical University, Dongguan, 523808, China
| | - Honghui Guo
- Department of Nutrition, School of Public Health, Guangdong Medical University, Dongguan, 523808, China. .,Dongguan Key Laboratory of Environmental Medicine, Guangdong Medical University, Dongguan, 523808, China.
| |
Collapse
|
799
|
Sanyal AJ, Van Natta ML, Clark J, Neuschwander-Tetri BA, Diehl A, Dasarathy S, Loomba R, Chalasani N, Kowdley K, Hameed B, Wilson LA, Yates KP, Belt P, Lazo M, Kleiner DE, Behling C, Tonascia J. Prospective Study of Outcomes in Adults with Nonalcoholic Fatty Liver Disease. N Engl J Med 2021; 385:1559-1569. [PMID: 34670043 PMCID: PMC8881985 DOI: 10.1056/nejmoa2029349] [Citation(s) in RCA: 618] [Impact Index Per Article: 154.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND The prognoses with respect to mortality and hepatic and nonhepatic outcomes across the histologic spectrum of nonalcoholic fatty liver disease (NAFLD) are not well defined. METHODS We prospectively followed a multicenter patient population that included the full histologic spectrum of NAFLD. The incidences of death and other outcomes were compared across baseline histologic characteristics. RESULTS A total of 1773 adults with NAFLD were followed for a median of 4 years. All-cause mortality increased with increasing fibrosis stages (0.32 deaths per 100 person-years for stage F0 to F2 [no, mild, or moderate fibrosis], 0.89 deaths per 100 persons-years for stage F3 [bridging fibrosis], and 1.76 deaths per 100 person-years for stage F4 [cirrhosis]). The incidence of liver-related complications per 100 person-years increased with fibrosis stage (F0 to F2 vs. F3 vs. F4) as follows: variceal hemorrhage (0.00 vs. 0.06 vs. 0.70), ascites (0.04 vs. 0.52 vs. 1.20), encephalopathy (0.02 vs. 0.75 vs. 2.39), and hepatocellular cancer (0.04 vs. 0.34 vs. 0.14). As compared with patients with stage F0 to F2 fibrosis, patients with stage F4 fibrosis also had a higher incidence of type 2 diabetes (7.53 vs. 4.45 events per 100 person-years) and a decrease of more than 40% in the estimated glomerular filtration rate (2.98 vs. 0.97 events per 100 person-years). The incidence of cardiac events and nonhepatic cancers were similar across fibrosis stages. After adjustment for age, sex, race, diabetes status, and baseline histologic severity, the incidence of any hepatic decompensation event (variceal hemorrhage, ascites, or encephalopathy) was associated with increased all-cause mortality (adjusted hazard ratio, 6.8; 95% confidence interval, 2.2 to 21.3). CONCLUSIONS In this prospective study involving patients with NAFLD, fibrosis stages F3 and F4 were associated with increased risks of liver-related complications and death. (Funded by the National Institute of Diabetes and Digestive and Kidney Diseases and others; NAFLD DB2 ClinicalTrials.gov number, NCT01030484.).
Collapse
Affiliation(s)
- Arun J Sanyal
- From the Virginia Commonwealth University School of Medicine, Richmond (A.J.S.); the Bloomberg School of Public Health, Johns Hopkins University, Baltimore (M.L.V.N., J.C., L.A.W., K.P.Y., P.B., M.L., J.T.), and the Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda (D.E.K.) - both in Maryland; Saint Louis University, St. Louis (B.A.N.-T.); Duke University, Durham, NC (A.M.D.); Cleveland Clinic, Cleveland (S.D.); the University of California, San Diego, School of Medicine, La Jolla (R.L., C.B.), and the University of California, San Francisco, School of Medicine, San Francisco (B.H.); Indiana University School of Medicine, Indianapolis (N.C.); and the Liver Institute Northwest, Seattle (K.K.)
| | - Mark L Van Natta
- From the Virginia Commonwealth University School of Medicine, Richmond (A.J.S.); the Bloomberg School of Public Health, Johns Hopkins University, Baltimore (M.L.V.N., J.C., L.A.W., K.P.Y., P.B., M.L., J.T.), and the Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda (D.E.K.) - both in Maryland; Saint Louis University, St. Louis (B.A.N.-T.); Duke University, Durham, NC (A.M.D.); Cleveland Clinic, Cleveland (S.D.); the University of California, San Diego, School of Medicine, La Jolla (R.L., C.B.), and the University of California, San Francisco, School of Medicine, San Francisco (B.H.); Indiana University School of Medicine, Indianapolis (N.C.); and the Liver Institute Northwest, Seattle (K.K.)
| | - Jeanne Clark
- From the Virginia Commonwealth University School of Medicine, Richmond (A.J.S.); the Bloomberg School of Public Health, Johns Hopkins University, Baltimore (M.L.V.N., J.C., L.A.W., K.P.Y., P.B., M.L., J.T.), and the Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda (D.E.K.) - both in Maryland; Saint Louis University, St. Louis (B.A.N.-T.); Duke University, Durham, NC (A.M.D.); Cleveland Clinic, Cleveland (S.D.); the University of California, San Diego, School of Medicine, La Jolla (R.L., C.B.), and the University of California, San Francisco, School of Medicine, San Francisco (B.H.); Indiana University School of Medicine, Indianapolis (N.C.); and the Liver Institute Northwest, Seattle (K.K.)
| | - Brent A Neuschwander-Tetri
- From the Virginia Commonwealth University School of Medicine, Richmond (A.J.S.); the Bloomberg School of Public Health, Johns Hopkins University, Baltimore (M.L.V.N., J.C., L.A.W., K.P.Y., P.B., M.L., J.T.), and the Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda (D.E.K.) - both in Maryland; Saint Louis University, St. Louis (B.A.N.-T.); Duke University, Durham, NC (A.M.D.); Cleveland Clinic, Cleveland (S.D.); the University of California, San Diego, School of Medicine, La Jolla (R.L., C.B.), and the University of California, San Francisco, School of Medicine, San Francisco (B.H.); Indiana University School of Medicine, Indianapolis (N.C.); and the Liver Institute Northwest, Seattle (K.K.)
| | - AnnaMae Diehl
- From the Virginia Commonwealth University School of Medicine, Richmond (A.J.S.); the Bloomberg School of Public Health, Johns Hopkins University, Baltimore (M.L.V.N., J.C., L.A.W., K.P.Y., P.B., M.L., J.T.), and the Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda (D.E.K.) - both in Maryland; Saint Louis University, St. Louis (B.A.N.-T.); Duke University, Durham, NC (A.M.D.); Cleveland Clinic, Cleveland (S.D.); the University of California, San Diego, School of Medicine, La Jolla (R.L., C.B.), and the University of California, San Francisco, School of Medicine, San Francisco (B.H.); Indiana University School of Medicine, Indianapolis (N.C.); and the Liver Institute Northwest, Seattle (K.K.)
| | - Srinivasan Dasarathy
- From the Virginia Commonwealth University School of Medicine, Richmond (A.J.S.); the Bloomberg School of Public Health, Johns Hopkins University, Baltimore (M.L.V.N., J.C., L.A.W., K.P.Y., P.B., M.L., J.T.), and the Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda (D.E.K.) - both in Maryland; Saint Louis University, St. Louis (B.A.N.-T.); Duke University, Durham, NC (A.M.D.); Cleveland Clinic, Cleveland (S.D.); the University of California, San Diego, School of Medicine, La Jolla (R.L., C.B.), and the University of California, San Francisco, School of Medicine, San Francisco (B.H.); Indiana University School of Medicine, Indianapolis (N.C.); and the Liver Institute Northwest, Seattle (K.K.)
| | - Rohit Loomba
- From the Virginia Commonwealth University School of Medicine, Richmond (A.J.S.); the Bloomberg School of Public Health, Johns Hopkins University, Baltimore (M.L.V.N., J.C., L.A.W., K.P.Y., P.B., M.L., J.T.), and the Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda (D.E.K.) - both in Maryland; Saint Louis University, St. Louis (B.A.N.-T.); Duke University, Durham, NC (A.M.D.); Cleveland Clinic, Cleveland (S.D.); the University of California, San Diego, School of Medicine, La Jolla (R.L., C.B.), and the University of California, San Francisco, School of Medicine, San Francisco (B.H.); Indiana University School of Medicine, Indianapolis (N.C.); and the Liver Institute Northwest, Seattle (K.K.)
| | - Naga Chalasani
- From the Virginia Commonwealth University School of Medicine, Richmond (A.J.S.); the Bloomberg School of Public Health, Johns Hopkins University, Baltimore (M.L.V.N., J.C., L.A.W., K.P.Y., P.B., M.L., J.T.), and the Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda (D.E.K.) - both in Maryland; Saint Louis University, St. Louis (B.A.N.-T.); Duke University, Durham, NC (A.M.D.); Cleveland Clinic, Cleveland (S.D.); the University of California, San Diego, School of Medicine, La Jolla (R.L., C.B.), and the University of California, San Francisco, School of Medicine, San Francisco (B.H.); Indiana University School of Medicine, Indianapolis (N.C.); and the Liver Institute Northwest, Seattle (K.K.)
| | - Kris Kowdley
- From the Virginia Commonwealth University School of Medicine, Richmond (A.J.S.); the Bloomberg School of Public Health, Johns Hopkins University, Baltimore (M.L.V.N., J.C., L.A.W., K.P.Y., P.B., M.L., J.T.), and the Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda (D.E.K.) - both in Maryland; Saint Louis University, St. Louis (B.A.N.-T.); Duke University, Durham, NC (A.M.D.); Cleveland Clinic, Cleveland (S.D.); the University of California, San Diego, School of Medicine, La Jolla (R.L., C.B.), and the University of California, San Francisco, School of Medicine, San Francisco (B.H.); Indiana University School of Medicine, Indianapolis (N.C.); and the Liver Institute Northwest, Seattle (K.K.)
| | - Bilal Hameed
- From the Virginia Commonwealth University School of Medicine, Richmond (A.J.S.); the Bloomberg School of Public Health, Johns Hopkins University, Baltimore (M.L.V.N., J.C., L.A.W., K.P.Y., P.B., M.L., J.T.), and the Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda (D.E.K.) - both in Maryland; Saint Louis University, St. Louis (B.A.N.-T.); Duke University, Durham, NC (A.M.D.); Cleveland Clinic, Cleveland (S.D.); the University of California, San Diego, School of Medicine, La Jolla (R.L., C.B.), and the University of California, San Francisco, School of Medicine, San Francisco (B.H.); Indiana University School of Medicine, Indianapolis (N.C.); and the Liver Institute Northwest, Seattle (K.K.)
| | - Laura A Wilson
- From the Virginia Commonwealth University School of Medicine, Richmond (A.J.S.); the Bloomberg School of Public Health, Johns Hopkins University, Baltimore (M.L.V.N., J.C., L.A.W., K.P.Y., P.B., M.L., J.T.), and the Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda (D.E.K.) - both in Maryland; Saint Louis University, St. Louis (B.A.N.-T.); Duke University, Durham, NC (A.M.D.); Cleveland Clinic, Cleveland (S.D.); the University of California, San Diego, School of Medicine, La Jolla (R.L., C.B.), and the University of California, San Francisco, School of Medicine, San Francisco (B.H.); Indiana University School of Medicine, Indianapolis (N.C.); and the Liver Institute Northwest, Seattle (K.K.)
| | - Katherine P Yates
- From the Virginia Commonwealth University School of Medicine, Richmond (A.J.S.); the Bloomberg School of Public Health, Johns Hopkins University, Baltimore (M.L.V.N., J.C., L.A.W., K.P.Y., P.B., M.L., J.T.), and the Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda (D.E.K.) - both in Maryland; Saint Louis University, St. Louis (B.A.N.-T.); Duke University, Durham, NC (A.M.D.); Cleveland Clinic, Cleveland (S.D.); the University of California, San Diego, School of Medicine, La Jolla (R.L., C.B.), and the University of California, San Francisco, School of Medicine, San Francisco (B.H.); Indiana University School of Medicine, Indianapolis (N.C.); and the Liver Institute Northwest, Seattle (K.K.)
| | - Patricia Belt
- From the Virginia Commonwealth University School of Medicine, Richmond (A.J.S.); the Bloomberg School of Public Health, Johns Hopkins University, Baltimore (M.L.V.N., J.C., L.A.W., K.P.Y., P.B., M.L., J.T.), and the Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda (D.E.K.) - both in Maryland; Saint Louis University, St. Louis (B.A.N.-T.); Duke University, Durham, NC (A.M.D.); Cleveland Clinic, Cleveland (S.D.); the University of California, San Diego, School of Medicine, La Jolla (R.L., C.B.), and the University of California, San Francisco, School of Medicine, San Francisco (B.H.); Indiana University School of Medicine, Indianapolis (N.C.); and the Liver Institute Northwest, Seattle (K.K.)
| | - Mariana Lazo
- From the Virginia Commonwealth University School of Medicine, Richmond (A.J.S.); the Bloomberg School of Public Health, Johns Hopkins University, Baltimore (M.L.V.N., J.C., L.A.W., K.P.Y., P.B., M.L., J.T.), and the Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda (D.E.K.) - both in Maryland; Saint Louis University, St. Louis (B.A.N.-T.); Duke University, Durham, NC (A.M.D.); Cleveland Clinic, Cleveland (S.D.); the University of California, San Diego, School of Medicine, La Jolla (R.L., C.B.), and the University of California, San Francisco, School of Medicine, San Francisco (B.H.); Indiana University School of Medicine, Indianapolis (N.C.); and the Liver Institute Northwest, Seattle (K.K.)
| | - David E Kleiner
- From the Virginia Commonwealth University School of Medicine, Richmond (A.J.S.); the Bloomberg School of Public Health, Johns Hopkins University, Baltimore (M.L.V.N., J.C., L.A.W., K.P.Y., P.B., M.L., J.T.), and the Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda (D.E.K.) - both in Maryland; Saint Louis University, St. Louis (B.A.N.-T.); Duke University, Durham, NC (A.M.D.); Cleveland Clinic, Cleveland (S.D.); the University of California, San Diego, School of Medicine, La Jolla (R.L., C.B.), and the University of California, San Francisco, School of Medicine, San Francisco (B.H.); Indiana University School of Medicine, Indianapolis (N.C.); and the Liver Institute Northwest, Seattle (K.K.)
| | - Cynthia Behling
- From the Virginia Commonwealth University School of Medicine, Richmond (A.J.S.); the Bloomberg School of Public Health, Johns Hopkins University, Baltimore (M.L.V.N., J.C., L.A.W., K.P.Y., P.B., M.L., J.T.), and the Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda (D.E.K.) - both in Maryland; Saint Louis University, St. Louis (B.A.N.-T.); Duke University, Durham, NC (A.M.D.); Cleveland Clinic, Cleveland (S.D.); the University of California, San Diego, School of Medicine, La Jolla (R.L., C.B.), and the University of California, San Francisco, School of Medicine, San Francisco (B.H.); Indiana University School of Medicine, Indianapolis (N.C.); and the Liver Institute Northwest, Seattle (K.K.)
| | - James Tonascia
- From the Virginia Commonwealth University School of Medicine, Richmond (A.J.S.); the Bloomberg School of Public Health, Johns Hopkins University, Baltimore (M.L.V.N., J.C., L.A.W., K.P.Y., P.B., M.L., J.T.), and the Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda (D.E.K.) - both in Maryland; Saint Louis University, St. Louis (B.A.N.-T.); Duke University, Durham, NC (A.M.D.); Cleveland Clinic, Cleveland (S.D.); the University of California, San Diego, School of Medicine, La Jolla (R.L., C.B.), and the University of California, San Francisco, School of Medicine, San Francisco (B.H.); Indiana University School of Medicine, Indianapolis (N.C.); and the Liver Institute Northwest, Seattle (K.K.)
| |
Collapse
|
800
|
Um YJ, Chang Y, Jung HS, Cho IY, Shin JH, Shin H, Wild SH, Byrne CD, Ryu S. Sleep Duration, Sleep Quality, and the Development of Nonalcoholic Fatty Liver Disease: A Cohort Study. Clin Transl Gastroenterol 2021; 12:e00417. [PMID: 34665792 PMCID: PMC8528229 DOI: 10.14309/ctg.0000000000000417] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 08/22/2021] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION The longitudinal relationship between sleep duration, sleep quality, and the risk of nonalcoholic fatty liver disease (NAFLD) is unknown. We aimed to examine the association between sleep duration, sleep quality, and NAFLD development. METHODS Using the Pittsburgh Sleep Quality Index, sleep duration and quality were evaluated for 143,306 NAFLD-free Korean adults with a mean age of 36.6 years, who were followed for an average of 4.0 years. Hepatic steatosis (HS) was assessed using ultrasonography and liver fibrosis by the fibrosis-4 index (FIB-4) or the NAFLD fibrosis score. Flexible parametric proportional hazard models were used to determine the hazard ratios (HRs) and 95% confidence intervals. RESULTS There were 27,817 subjects with incident HS, of whom 1,471 had incident HS plus intermediate/high FIB-4. Multivariable-adjusted HRs (95% confidence intervals) for incident HS comparing sleep durations of ≤5, 6, 8, and ≥ 9 hours with 7 hours were 1.19 (1.14-1.23), 1.07 (1.04-1.10), 0.98 (0.94-1.02), and 0.95 (0.87-1.03), respectively. The corresponding HRs for incident HS plus intermediate/high FIB-4 were 1.30 (1.11-1.54), 1.14 (1.01-1.29), 1.11 (0.93-1.33), and 1.08 (0.71-1.63). The association between sleep duration and HS plus intermediate/high FIB-4 was inverse in individuals with good sleep quality but tended to be U-shaped in those with poor sleep quality. The results were similar if FIB-4 was replaced by the NAFLD fibrosis score. DISCUSSION In young adults, short sleep duration was independently associated with an increased risk of incident NAFLD with or without intermediate/high fibrosis score, suggesting a role for inadequate sleep quantity in NAFLD risk and severity.
Collapse
Affiliation(s)
- Yoo Jin Um
- Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, KR, South Korea
| | - Yoosoo Chang
- Center for Cohort Studies, Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, KR, South Korea
- Department of Occupational and Environmental Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, KR, South Korea
- Department of Clinical Research Design and Evaluation, SAIHST, Sungkyunkwan University, Seoul, KR, South Korea
| | - Hyun-Suk Jung
- Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, KR, South Korea
- Center for Cohort Studies, Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, KR, South Korea
| | - In Young Cho
- Department of Family Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, KR, South Korea
| | - Jun Ho Shin
- Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, KR, South Korea
- Department of Surgery, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, KR, South Korea
| | - Hocheol Shin
- Center for Cohort Studies, Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, KR, South Korea
- Department of Family Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, KR, South Korea
| | - Sarah H. Wild
- Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Christopher D. Byrne
- Nutrition and Metabolism, Faculty of Medicine, University of Southampton, Southampton, UK
- National Institute for Health Research Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Seungho Ryu
- Center for Cohort Studies, Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, KR, South Korea
- Department of Occupational and Environmental Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, KR, South Korea
- Department of Clinical Research Design and Evaluation, SAIHST, Sungkyunkwan University, Seoul, KR, South Korea
| |
Collapse
|