101
|
Avignon A. Protecting the Liver: Should We Substitute Fruit Juices for Sugar-Sweetened Beverages? Diabetes Care 2022; 45:1032-1034. [PMID: 35561135 DOI: 10.2337/dci22-0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
102
|
Wu H, He X, Li Q, Zheng Y, Rayamajhi S, Thapa A, Meng G, Zhang Q, Liu L, Wu H, Gu Y, Zhang S, Zhang T, Wang X, Zhang J, Cao Z, Dong J, Zheng X, Zhang X, Dong X, Sun S, Wang X, Zhou M, Jia Q, Song K, Chang H, Niu K. Relationship between the consumption of wholegrain and nonalcoholic fatty liver disease: The TCLSIH cohort study. Clin Nutr 2022; 41:1483-1490. [DOI: 10.1016/j.clnu.2022.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 05/04/2022] [Accepted: 05/13/2022] [Indexed: 11/03/2022]
|
103
|
Wen Y, Chen C, Kong X, Xia Z, Kong W, Si K, Han P, Vivian Liu W, Li X. Pancreatic fat infiltration, β-cell function and insulin resistance: A study of the young patients with obesity. Diabetes Res Clin Pract 2022; 187:109860. [PMID: 35367311 DOI: 10.1016/j.diabres.2022.109860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 03/07/2022] [Accepted: 03/29/2022] [Indexed: 11/25/2022]
Abstract
OBJECTIVE This study aimed to investigate the relationship between pancreatic fat infiltration (PFI) and glucose metabolism disorder, β-cell function and insulin resistance in patients with obesity. METHODS Pancreatic fat fraction (PFF) was quantified by MRI IDEAL-IQ technique. PFF greater than 6.2 % was defined as PFI, and 34 obese patients were divided into PFI and non-PFI groups. The 5-point plasma glucose and insulin values during oral glucose tolerance test (OGTT) were recorded. OGTT-derived indices of insulin resistance and β-cell function were calculated. RESULTS Glucose values levels at 0-120 min during OGTT were significantly higher and β-cell function variables were lower in PFI group than non-PFI group. While indices of insulin resistance were not significantly different between two groups. Correlation analysis showed that PFF was positively correlated with glucose levels at 0, 30 and 60 min, negatively correlated with β-cell function variables and not significantly correlated with indices of insulin resistance. However, these associations of PFF with β-cell function and glucose levels were only present in type 2 diabetes mellitus (T2DM) group but not in non-T2DM group. CONCLUSION There is an association between PFI and impaired β-cell function, and increased pancreatic fat may be a potential risk factor for the development of T2DM.
Collapse
Affiliation(s)
- Yu Wen
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Cen Chen
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Xiangchuang Kong
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Zefeng Xia
- Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wen Kong
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Keke Si
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Ping Han
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | | | - Xin Li
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China.
| |
Collapse
|
104
|
Ciardullo S, Mantovani A, Ciaccio A, Carbone M, Invernizzi P, Perseghin G. Hepatitis C virus infection and diabetes: A complex bidirectional relationship. Diabetes Res Clin Pract 2022; 187:109870. [PMID: 35398458 DOI: 10.1016/j.diabres.2022.109870] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/22/2022] [Accepted: 04/04/2022] [Indexed: 11/03/2022]
Abstract
Chronic hepatitis C (CHC) and diabetes represent two severe chronic conditions responsible for a considerable number of deaths worldwide. They have a complex, bidirectional relationship. On the one hand, several cohort studies have shown that chronic HCV infection increases both the risk of developing diabetes in non-diabetic subjects (by inducing insulin resistance and promoting β-cell dysfunction) as well as the risk of developing macro and microvascular complications in patients with known diabetes; on the other hand, diabetes is an independent risk factor for liver-related events among patients with CHC, including a higher incidence of hepatocellular carcinoma, liver-related death and transplantation. Importantly, sustained virological response, which can be obtained in the vast majority of patients with the use of direct antiviral agents, does not only lead to a lower rate of liver-related outcomes, but also to improvements of glycemic control and reduction in the rate of complications among patients with diabetes. The aim of this review is to summarize available clinical evidence on the association among CHC, diabetes and related clinical outcomes. We will also briefly discuss the biological mechanisms underpinning the association between CHC and diabetes, as well as the implications this relationship should have on everyday clinical practice.
Collapse
Affiliation(s)
- Stefano Ciardullo
- Department of Medicine and Rehabilitation, Monza Policlinico di Monza, Monza, Italy; Department of Medicine and Surgery, University of Milan Bicocca, Monza, Italy.
| | - Alessandro Mantovani
- Section of Endocrinology, Diabetes and Metabolic Diseases, Department of Medicine, University of Verona
| | - Antonio Ciaccio
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy; European Reference Network on Hepatological Diseases (ERN RARE-LIVER), San Gerardo Hospital, Monza, Italy
| | - Marco Carbone
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy; European Reference Network on Hepatological Diseases (ERN RARE-LIVER), San Gerardo Hospital, Monza, Italy
| | - Pietro Invernizzi
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy; European Reference Network on Hepatological Diseases (ERN RARE-LIVER), San Gerardo Hospital, Monza, Italy
| | - Gianluca Perseghin
- Department of Medicine and Rehabilitation, Monza Policlinico di Monza, Monza, Italy; Department of Medicine and Surgery, University of Milan Bicocca, Monza, Italy
| |
Collapse
|
105
|
Higashiura Y, Tanaka M, Mori K, Mikami T, Hosaka I, Ohnishi H, Hanawa N, Furuhashi M. High fibrosis-4 index predicts the new onset of ischaemic heart disease during a 10-year period in a general population. EUROPEAN HEART JOURNAL OPEN 2022; 2:oeac030. [PMID: 35919342 PMCID: PMC9242084 DOI: 10.1093/ehjopen/oeac030] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 03/22/2022] [Accepted: 04/11/2022] [Indexed: 12/13/2022]
Abstract
Aims The fibrosis-4 (FIB-4) index, calculated using age, platelet count, and levels of aspartate aminotransferase and alanine aminotransferase, is a non-invasive indicator for the detection of liver fibrosis. Advanced hepatic fibrosis is associated with morbidity and mortality in patients with non-alcoholic fatty liver disease. However, the relationship between liver fibrosis and the development of ischaemic heart disease (IHD) has not fully been addressed. Methods and results We investigated the association between the FIB-4 index and the new onset of IHD during a 10-year period in a general population of subjects who received annual health examinations (n = 28 990). After exclusion of subjects with missing data and those with a history of IHD at baseline, a total of 13 448 subjects (men/women: 8774/4674, mean age: 48 years) were included. During the 10-year period, 378 men (4.3%) and 77 women (1.6%) had a new onset of IHD. Multivariable Cox proportional hazard models with a restricted cubic spline showed that hazard risk for the development of IHD increased with a higher FIB-4 index at baseline after adjustment of age, sex, fatty liver (FL) determined by ultrasonography, estimated glomerular filtration rate, habits of current smoking and alcohol drinking, family history of IHD, and diagnosis of hypertension, diabetes mellitus and dyslipidaemia. When divided by FL, the FIB-4 index becomes an independent predictor for the development of IHD in subjects with FL but not in those without FL. The addition of the FIB-4 index to traditional risk factors for IHD significantly improved the discriminatory capability. Conclusion A high level of the FIB-4 index predicts the new onset of IHD during a 10-year period.
Collapse
Affiliation(s)
- Yukimura Higashiura
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Marenao Tanaka
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kazuma Mori
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takuma Mikami
- Department of Cardiovascular Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Itaru Hosaka
- Department of Cardiovascular Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hirofumi Ohnishi
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Nagisa Hanawa
- Department of Health Checkup and Promotion, Keijinkai Maruyama Clinic, Sapporo, Japan
| | - Masato Furuhashi
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
106
|
Li N, Tan H, Xie A, Li C, Fu X, Xang W, Kirim A, Huang X. Value of the triglyceride glucose index combined with body mass index in identifying non-alcoholic fatty liver disease in patients with type 2 diabetes. BMC Endocr Disord 2022; 22:101. [PMID: 35428219 PMCID: PMC9011983 DOI: 10.1186/s12902-022-00993-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 03/16/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The triglyceride glucose index combined with body mass index is a new index that reflects the degree of insulin resistance. In this cross-sectional study, we aimed to explore the predictive value of the triglyceride glucose-body mass index (TyG-BMI) in relation to the occurrence of non-alcoholic fatty liver disease (NAFLD) in the Chinese population with type 2 diabetes (T2D). METHODS We selected 826 patients with T2D who were hospitalized at the Department of Endocrinology and Metabolism of Karamay People's Hospital from September 2016 to October 2018 for this research. The height, weight, fasting blood glucose, serum insulin, and lipid profiles of the subjects were collected. The liver ultrasound showed any degree of echogenic enhancement of liver tissue and the liver appeared brighter than the renal cortex on ultrasound were considered to be NAFLD. The logistic regression analysis was performed to estimate associations between the triglyceride glucose index (TyG), TyG-BMI index, insulin resistance index (HOMA-IR) and the ratio of the triglycerides to high-density lipoprotein-cholesterol with a diagnosis of NAFLD. The receiver operating characteristic curve method was used to analyze its predictive value for NAFLD. RESULTS Results of the logistic regression analysis showed that the odds ratios of NAFLD were 6.535 (3.70-11.53) and 4.868 (2.576-9.200) for the TyG-BMI before and after correction,respectively(P < 0.001). The area under the curve (AUC) for TyG-BMI was 0.727 (0.691-0.764), which was the highest among all the other parameters studied. CONCLUSION Compared with the TyG index, the TG/HDL-C and HOMA-IR, the TyG-BMI was a more effective predictor of NAFLD in T2D.
Collapse
Affiliation(s)
- Nong Li
- Department of Endocrinology and Metabolism, People's Hospital of Karamay, Xinjiang, China.
| | - Huiwen Tan
- Department of Endocrinology Metabolism, West China Hospital of Sichuan University, Chengdu, China
| | - Aixia Xie
- Department of Endocrinology and Metabolism, People's Hospital of Karamay, Xinjiang, China
| | - Cheng Li
- First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
| | - Xuan Fu
- Department of Endocrinology and Metabolism, People's Hospital of Karamay, Xinjiang, China
| | - Weiting Xang
- Department of Endocrinology and Metabolism, People's Hospital of Karamay, Xinjiang, China
| | - Amina Kirim
- Department of Endocrinology and Metabolism, People's Hospital of Karamay, Xinjiang, China
| | - Xuefang Huang
- Department of Endocrinology and Metabolism, People's Hospital of Karamay, Xinjiang, China.
| |
Collapse
|
107
|
Talenezhad N, Mirzavandi F, Rahimpour S, Amel Shahbaz AP, Mohammadi M, Hosseinzadeh M. Empirically derived dietary pattern and odds of non-alcoholic fatty liver diseases in overweight and obese adults: a case-control study. BMC Gastroenterol 2022; 22:158. [PMID: 35354433 PMCID: PMC8966273 DOI: 10.1186/s12876-022-02222-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 03/17/2022] [Indexed: 12/26/2022] Open
Abstract
Background The prevalence of non-alcoholic fatty liver disease (NAFLD) is rising at an exponential rate throughout the world. Given the confirmed association between nutritional status and NAFLD, this study aimed to investigate the relationship of dietary patterns with NAFLD in overweight and obese adults. Methods In this age- and gender-matched case–control study, 115 newly diagnosed cases and 102 control individuals participated. A validated 178-item semi-quantitative food frequency questionnaire was administered to assess the participants' dietary data. Dietary patterns were extracted from 24 predefined food groups by factor analysis. Multivariate logistic regression was run to evaluate the relationship between dietary patterns and NAFLD. Results Factor analysis resulted in: “western”, “traditional”, and “snack and sweets” dietary patterns. The NAFLD odds were greater in participants at the highest quintile of the “western” dietary pattern than the lowest quintile (OR: 3.52; 95% CI: 1.64, 8.61). A significant increasing trend was observed in NAFLD odds across increasing quintiles of the “western” dietary pattern (P-trend = 0.01). After adjusting for the potential confounders, this relationship remained significant (OR: 3.30; 95% CI: 1.06–10.27). After full adjustments, NAFLD had no association with “traditional” or “snack and sweets” dietary patterns. Conclusion The “western” dietary pattern containing fast food, refined grains, liquid oil, pickles, high-fat dairy, sweet desserts, red meat, tea, and coffee was associated with increased odds of NAFLD. However, further prospective studies are required to establish these results.
Collapse
Affiliation(s)
- Nasir Talenezhad
- Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Farhang Mirzavandi
- Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Shahab Rahimpour
- Gastroentrology Department, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Amir Pasha Amel Shahbaz
- Department of Radiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mohammad Mohammadi
- Department of Community Medicine, School of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Mahdieh Hosseinzadeh
- Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran. .,Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
108
|
Lustig RH, Collier D, Kassotis C, Roepke TA, Ji Kim M, Blanc E, Barouki R, Bansal A, Cave MC, Chatterjee S, Choudhury M, Gilbertson M, Lagadic-Gossmann D, Howard S, Lind L, Tomlinson CR, Vondracek J, Heindel JJ. Obesity I: Overview and molecular and biochemical mechanisms. Biochem Pharmacol 2022; 199:115012. [PMID: 35393120 PMCID: PMC9050949 DOI: 10.1016/j.bcp.2022.115012] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 03/12/2022] [Accepted: 03/15/2022] [Indexed: 02/06/2023]
Abstract
Obesity is a chronic, relapsing condition characterized by excess body fat. Its prevalence has increased globally since the 1970s, and the number of obese and overweight people is now greater than those underweight. Obesity is a multifactorial condition, and as such, many components contribute to its development and pathogenesis. This is the first of three companion reviews that consider obesity. This review focuses on the genetics, viruses, insulin resistance, inflammation, gut microbiome, and circadian rhythms that promote obesity, along with hormones, growth factors, and organs and tissues that control its development. It shows that the regulation of energy balance (intake vs. expenditure) relies on the interplay of a variety of hormones from adipose tissue, gastrointestinal tract, pancreas, liver, and brain. It details how integrating central neurotransmitters and peripheral metabolic signals (e.g., leptin, insulin, ghrelin, peptide YY3-36) is essential for controlling energy homeostasis and feeding behavior. It describes the distinct types of adipocytes and how fat cell development is controlled by hormones and growth factors acting via a variety of receptors, including peroxisome proliferator-activated receptor-gamma, retinoid X, insulin, estrogen, androgen, glucocorticoid, thyroid hormone, liver X, constitutive androstane, pregnane X, farnesoid, and aryl hydrocarbon receptors. Finally, it demonstrates that obesity likely has origins in utero. Understanding these biochemical drivers of adiposity and metabolic dysfunction throughout the life cycle lends plausibility and credence to the "obesogen hypothesis" (i.e., the importance of environmental chemicals that disrupt these receptors to promote adiposity or alter metabolism), elucidated more fully in the two companion reviews.
Collapse
Affiliation(s)
- Robert H Lustig
- Division of Endocrinology, Department of Pediatrics, University of California, San Francisco, CA 94143, United States
| | - David Collier
- Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States
| | - Christopher Kassotis
- Institute of Environmental Health Sciences and Department of Pharmacology, Wayne State University, Detroit, MI 48202, United States
| | - Troy A Roepke
- School of Environmental and Biological Sciences, Rutgers University, New Brunswick, NJ 08901, United States
| | - Min Ji Kim
- Department of Biochemistry and Toxicology, University of Paris, INSERM U1224 (T3S), 75006 Paris, France
| | - Etienne Blanc
- Department of Biochemistry and Toxicology, University of Paris, INSERM U1224 (T3S), 75006 Paris, France
| | - Robert Barouki
- Department of Biochemistry and Toxicology, University of Paris, INSERM U1224 (T3S), 75006 Paris, France
| | - Amita Bansal
- College of Health & Medicine, Australian National University, Canberra, Australia
| | - Matthew C Cave
- Division of Gastroenterology, Hepatology and Nutrition, University of Louisville, Louisville, KY 40402, United States
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, University of South Carolina, Columbia, SC 29208, United States
| | - Mahua Choudhury
- College of Pharmacy, Texas A&M University, College Station, TX 77843, United States
| | - Michael Gilbertson
- Occupational and Environmental Health Research Group, University of Stirling, Stirling, Scotland, United Kingdom
| | - Dominique Lagadic-Gossmann
- Research Institute for Environmental and Occupational Health, University of Rennes, INSERM, EHESP, Rennes, France
| | - Sarah Howard
- Healthy Environment and Endocrine Disruptor Strategies, Commonweal, Bolinas, CA 92924, United States
| | - Lars Lind
- Department of Medical Sciences, University of Uppsala, Uppsala, Sweden
| | - Craig R Tomlinson
- Norris Cotton Cancer Center, Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, United States
| | - Jan Vondracek
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Jerrold J Heindel
- Healthy Environment and Endocrine Disruptor Strategies, Commonweal, Bolinas, CA 92924, United States.
| |
Collapse
|
109
|
Qiao L, Men L, Yu S, Yao J, Li Y, Wang M, Yu Y, Wang N, Ran L, Wu Y, Du J. Hepatic deficiency of selenoprotein S exacerbates hepatic steatosis and insulin resistance. Cell Death Dis 2022; 13:275. [PMID: 35347118 PMCID: PMC8960781 DOI: 10.1038/s41419-022-04716-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 02/18/2022] [Accepted: 03/09/2022] [Indexed: 12/13/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is closely associated with insulin resistance (IR) and type 2 diabetes mellitus (T2DM), which are all complex metabolic disorders. Selenoprotein S (SelS) is an endoplasmic reticulum (ER) resident selenoprotein involved in regulating ER stress and has been found to participate in the occurrence and development of IR and T2DM. However, the potential role and mechanism of SelS in NAFLD remains unclear. Here, we analyzed SelS expression in the liver of high-fat diet (HFD)-fed mice and obese T2DM model (db/db) mice and generated hepatocyte-specific SelS knockout (SelSH-KO) mice using the Cre-loxP system. We showed that hepatic SelS expression levels were significantly downregulated in HFD-fed mice and db/db mice. Hepatic SelS deficiency markedly increased ER stress markers in the liver and caused hepatic steatosis via increased fatty acid uptake and reduced fatty acid oxidation. Impaired insulin signaling was detected in the liver of SelSH-KO mice with decreased phosphorylation levels of insulin receptor substrate 1 (IRS1) and protein kinase B (PKB/Akt), which ultimately led to disturbed glucose homeostasis. Meanwhile, our results showed hepatic protein kinase Cɛ (PKCɛ) activation participated in the negative regulation of insulin signaling in SelSH-KO mice. Moreover, the inhibitory effect of SelS on hepatic steatosis and IR was confirmed by SelS overexpression in primary hepatocytes in vitro. Thus, we conclude that hepatic SelS plays a key role in regulating hepatic lipid accumulation and insulin action, suggesting that SelS may be a potential intervention target for the prevention and treatment of NAFLD and T2DM.
Collapse
Affiliation(s)
- Lu Qiao
- Department of Endocrinology, the First Affiliated Hospital of Dalian Medical University, Dalian, China.,Dalian Key Laboratory of Prevention and Treatment of Metabolic Diseases and the Vascular Complications, Dalian, China
| | - Lili Men
- Department of Endocrinology, the First Affiliated Hospital of Dalian Medical University, Dalian, China.,Dalian Key Laboratory of Prevention and Treatment of Metabolic Diseases and the Vascular Complications, Dalian, China
| | - Shanshan Yu
- Department of Endocrinology, the First Affiliated Hospital of Dalian Medical University, Dalian, China.,Dalian Key Laboratory of Prevention and Treatment of Metabolic Diseases and the Vascular Complications, Dalian, China
| | - Junjie Yao
- Department of Endocrinology, the First Affiliated Hospital of Dalian Medical University, Dalian, China.,Dalian Key Laboratory of Prevention and Treatment of Metabolic Diseases and the Vascular Complications, Dalian, China
| | - Yu Li
- Department of Endocrinology, the First Affiliated Hospital of Dalian Medical University, Dalian, China.,Dalian Key Laboratory of Prevention and Treatment of Metabolic Diseases and the Vascular Complications, Dalian, China
| | - Mingming Wang
- Department of Endocrinology, the First Affiliated Hospital of Dalian Medical University, Dalian, China.,Dalian Key Laboratory of Prevention and Treatment of Metabolic Diseases and the Vascular Complications, Dalian, China
| | - Ying Yu
- Department of Endocrinology, the First Affiliated Hospital of Dalian Medical University, Dalian, China.,Dalian Key Laboratory of Prevention and Treatment of Metabolic Diseases and the Vascular Complications, Dalian, China
| | - Ning Wang
- Institute for Genome Engineered Animal Models of Human Diseases, Dalian Medical University, Dalian, China.,National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, China
| | - Liyuan Ran
- Institute for Genome Engineered Animal Models of Human Diseases, Dalian Medical University, Dalian, China.,National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, China
| | - Yingjie Wu
- Institute for Genome Engineered Animal Models of Human Diseases, Dalian Medical University, Dalian, China. .,National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, China. .,Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, USA.
| | - Jianling Du
- Department of Endocrinology, the First Affiliated Hospital of Dalian Medical University, Dalian, China. .,Dalian Key Laboratory of Prevention and Treatment of Metabolic Diseases and the Vascular Complications, Dalian, China.
| |
Collapse
|
110
|
Mahmoudi A, Butler AE, Majeed M, Banach M, Sahebkar A. Investigation of the Effect of Curcumin on Protein Targets in NAFLD Using Bioinformatic Analysis. Nutrients 2022; 14:nu14071331. [PMID: 35405942 PMCID: PMC9002953 DOI: 10.3390/nu14071331] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/15/2022] [Accepted: 03/21/2022] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND: Non-alcoholic fatty liver disease (NAFLD) is a prevalent metabolic disorder. Defects in function/expression of genes/proteins are critical in initiation/progression of NAFLD. Natural products may modulate these genes/proteins. Curcumin improves steatosis, inflammation, and fibrosis progression. Here, bioinformatic tools, gene−drug and gene-disease databases were utilized to explore targets, interactions, and pathways through which curcumin could impact NAFLD. METHODS: Significant curcumin−protein interaction was identified (high-confidence:0.7) in the STITCH database. Identified proteins were investigated to determine association with NAFLD. gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were analyzed for significantly involved targets (p < 0.01). Specificity of obtained targets with NAFLD was estimated and investigated in Tissue/Cells−gene associations (PanglaoDB Augmented 2021, Mouse Gene Atlas) and Disease−gene association-based EnrichR algorithms (Jensen DISEASES, DisGeNET). RESULTS: Two collections were constructed: 227 protein−curcumin interactions and 95 NAFLD-associated genes. By Venn diagram, 14 significant targets were identified, and their biological pathways evaluated. Based on gene ontology, most targets involved stress and lipid metabolism. KEGG revealed chemical carcinogenesis, the AGE-RAGE signaling pathway in diabetic complications and NAFLD as the most common significant pathways. Specificity to diseases database (EnrichR algorithm) revealed specificity for steatosis/steatohepatitis. CONCLUSION: Curcumin may improve, or inhibit, progression of NAFLD through activation/inhibition of NAFLD-related genes.
Collapse
Affiliation(s)
- Ali Mahmoudi
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad 9177899191, Iran;
| | - Alexandra E. Butler
- Research Department, Royal College of Surgeons in Ireland Bahrain, Adliya 15503, Bahrain;
| | | | - Maciej Banach
- Nephrology and Hypertension, Department of Preventive Cardiology and Lipidology, Medical University of Lodz, 93-338 Lodz, Poland
- Cardiovascular Research Centre, University of Zielona Gora, 65-417 Zielona Gora, Poland
- Correspondence: (M.B.); (A.S.)
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9177899191, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad 9177899191, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177899191, Iran
- Correspondence: (M.B.); (A.S.)
| |
Collapse
|
111
|
Nguyen MT, Min KH, Lee W. MiR-183-5p Induced by Saturated Fatty Acids Hinders Insulin Signaling by Downregulating IRS-1 in Hepatocytes. Int J Mol Sci 2022; 23:ijms23062979. [PMID: 35328400 PMCID: PMC8953084 DOI: 10.3390/ijms23062979] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/07/2022] [Accepted: 03/09/2022] [Indexed: 02/06/2023] Open
Abstract
Excessive saturated fatty acids (SFA) uptake is known to be a primary cause of obesity, a widely acknowledged risk factor of insulin resistance and type 2 diabetes. Although specific microRNAs (miRNAs) targeting insulin signaling intermediates are dysregulated by SFA, their effects on insulin signaling and sensitivity are largely unknown. Here, we investigated the role of SFA-induced miR-183-5p in the regulation of proximal insulin signaling molecules and the development of hepatic insulin resistance. HepG2 hepatocytes treated with palmitate and the livers of high-fat diet (HFD)-fed mice exhibited impaired insulin signaling resulting from dramatic reductions in the protein expressions of insulin receptor (INSR) and insulin receptor substrate-1 (IRS-1). Differential expression analysis showed the level of miR-183-5p, which tentatively targets the 3'UTR of IRS-1, was significantly elevated in palmitate-treated HepG2 hepatocytes and the livers of HFD-fed mice. Dual-luciferase analysis showed miR-183-5p bound directly to the 3'UTR of IRS-1 and reduced IRS-1 expression at the post-transcriptional stage. Moreover, transfection of HepG2 hepatocytes with miR-183-5p mimic significantly inhibited IRS-1 expression and hindered insulin signaling, consequently inhibiting insulin-stimulated glycogen synthesis. Collectively, this study reveals a novel mechanism whereby miR-183-5p induction by SFA impairs insulin signaling and suggests miR-183-5p plays a crucial role in the pathogenesis of hepatic insulin resistance in the background of obesity.
Collapse
Affiliation(s)
- Mai Thi Nguyen
- Department of Biochemistry, College of Medicine, Dongguk University, 123 Dongdae-ro, Gyeongju 38066, Korea; (M.T.N.); (K.-H.M.)
| | - Kyung-Ho Min
- Department of Biochemistry, College of Medicine, Dongguk University, 123 Dongdae-ro, Gyeongju 38066, Korea; (M.T.N.); (K.-H.M.)
| | - Wan Lee
- Department of Biochemistry, College of Medicine, Dongguk University, 123 Dongdae-ro, Gyeongju 38066, Korea; (M.T.N.); (K.-H.M.)
- Channelopathy Research Center, College of Medicine, Dongguk University, 32 Dongguk-ro, Ilsan Dong-gu, Goyang 10326, Korea
- Correspondence: ; Tel.: +82-54-770-2409
| |
Collapse
|
112
|
Pang Y, Kartsonaki C, Lv J, Millwood IY, Fairhurst-Hunter Z, Turnbull I, Bragg F, Hill MR, Yu C, Guo Y, Chen Y, Yang L, Clarke R, Walters RG, Wu M, Chen J, Li L, Chen Z, Holmes MV. Adiposity, metabolomic biomarkers, and risk of nonalcoholic fatty liver disease: a case-cohort study. Am J Clin Nutr 2022; 115:799-810. [PMID: 34902008 PMCID: PMC8895224 DOI: 10.1093/ajcn/nqab392] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/06/2021] [Accepted: 11/18/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Globally, the burden of obesity and associated nonalcoholic fatty liver disease (NAFLD) are rising, but little is known about the role that circulating metabolomic biomarkers play in mediating their association. OBJECTIVES We aimed to examine the observational and genetic associations of adiposity with metabolomic biomarkers and the observational associations of metabolomic biomarkers with incident NAFLD. METHODS A case-subcohort study within the prospective China Kadoorie Biobank included 176 NAFLD cases and 180 subcohort individuals and measured 1208 metabolites in stored baseline plasma using a Metabolon assay. In the subcohort the observational and genetic associations of BMI with biomarkers were assessed using linear regression, with adjustment for multiple testing. Cox regression was used to estimate adjusted HRs for NAFLD associated with biomarkers. RESULTS In observational analyses, BMI (kg/m2; mean: 23.9 in the subcohort) was associated with 199 metabolites at a 5% false discovery rate. The effects of genetically elevated BMI with specific metabolites were directionally consistent with the observational associations. Overall, 35 metabolites were associated with NAFLD risk, of which 15 were also associated with BMI, including glutamate (HR per 1-SD higher metabolite: 1.95; 95% CI: 1.48, 2.56), cysteine-glutathione disulfide (0.44; 0.31, 0.62), diaclyglycerol (C32:1) (1.71; 1.24, 2.35), behenoyl dihydrosphingomyelin (C40:0) (1.92; 1.42, 2.59), butyrylcarnitine (C4) (1.91; 1.38, 2.35), 2-hydroxybehenate (1.81; 1.34, 2.45), and 4-cholesten-3-one (1.79; 1.27, 2.54). The discriminatory performance of known risk factors was increased when 28 metabolites were also considered simultaneously in the model (weighted C-statistic: 0.84 to 0.90; P < 0.001). CONCLUSIONS Among relatively lean Chinese adults, a range of metabolomic biomarkers are associated with NAFLD risk and these biomarkers may lie on the pathway between adiposity and NAFLD.
Collapse
Affiliation(s)
- Yuanjie Pang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Christiana Kartsonaki
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
- Medical Research Council Population Health Research Unit (MRC PHRU) at the University of Oxford, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Jun Lv
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness and Response (PKU-PHEPR), Peking University, Beijing, China
| | - Iona Y Millwood
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
- Medical Research Council Population Health Research Unit (MRC PHRU) at the University of Oxford, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Zammy Fairhurst-Hunter
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Iain Turnbull
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Fiona Bragg
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
- Medical Research Council Population Health Research Unit (MRC PHRU) at the University of Oxford, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Michael R Hill
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Canqing Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness and Response (PKU-PHEPR), Peking University, Beijing, China
| | - Yu Guo
- Chinese Academy of Medical Sciences, Beijing, China
| | - Yiping Chen
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
- Medical Research Council Population Health Research Unit (MRC PHRU) at the University of Oxford, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Ling Yang
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
- Medical Research Council Population Health Research Unit (MRC PHRU) at the University of Oxford, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Robert Clarke
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Robin G Walters
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
- Medical Research Council Population Health Research Unit (MRC PHRU) at the University of Oxford, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Ming Wu
- Jiangsu Center for Disease Control and Prevention, Nanjing, China
| | - Junshi Chen
- National Center for Food Safety Risk Assessment, Beijing, China
| | - Liming Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness and Response (PKU-PHEPR), Peking University, Beijing, China
| | - Zhengming Chen
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
- Medical Research Council Population Health Research Unit (MRC PHRU) at the University of Oxford, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Michael V Holmes
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
- Medical Research Council Population Health Research Unit (MRC PHRU) at the University of Oxford, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
- National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospital, Oxford, United Kingdom
| |
Collapse
|
113
|
Ren Q, Wang H, Zeng Y, Fang X, Wang M, Li D, Huang W, Xu Y. Circulating chemerin levels in metabolic-associated fatty liver disease: a systematic review and meta-analysis. Lipids Health Dis 2022; 21:27. [PMID: 35236351 PMCID: PMC8889738 DOI: 10.1186/s12944-022-01637-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/17/2022] [Indexed: 12/21/2022] Open
Abstract
Background and objectives Chemerin is a brand-new adipokine that has been linked to both inflammation and metabolic dysfunction. Even though a rising number of studies have connected chemerin to metabolic-associated fatty liver disease (MAFLD), formerly referred to as non-alcoholic fatty liver disease (NAFLD), this association has been controversial. Methods A comprehensive literature search was undertaken up to February 1, 2022, in the PubMed, Embase, Web of Science, CNKI, WANFANG, and CBM library databases. Circulating chemerin levels were obtained and summarized using the standardized mean difference (SMD) and 95% confidence interval (CI). Subgroup and meta-regression analyses were conducted to examine the possibility of heterogeneity. Results A total of 17 studies involving 2580 participants (1584 MAFLD patients and 996 controls) evaluated circulating chemerin levels in patients with MAFLD. The present study showed that higher chemerin levels were found in patients with MAFLD (SMD: 1.32; 95% CI: 0.29, 2.35) and nonalcoholic fatty liver (NAFL) (SMD: 0.75; 95% CI: 0.01, 1.50) compared to controls. However, circulating chemerin levels did not differ significantly in the following comparisons: nonalcoholic steatohepatitis (NASH) patients and controls (SMD: 0.75; 95% CI: -0.52, 2.03); NASH patients and NAFL patients (SMD: 0.16; 95% CI: -0.39, 0.70); moderate to severe steatosis and mild steatosis (SMD: 0.55; 95% CI: -0.59, 1.69); present liver fibrosis and absent liver fibrosis (SMD: 0.66; 95% CI: -0.42, 1.74); present lobular inflammation and absent lobular inflammation (SMD: 0.45; 95% CI: -0.53, 1.42); and present portal inflammation and absent portal inflammation (SMD: 1.92; 95% CI: -0.85, 4.69). Conclusions Chemerin levels were considerably greater in patients with MAFLD than in controls, despite the fact that they were not significantly linked to different liver tissue lesions of MAFLD. In different subtypes of MAFLD, in comparison to healthy controls, the chemerin levels of NAFL patients were higher, whereas, there was no obvious difference in chemerin levels between NASH patients and controls. It is possible that chemerin will be used as a biomarker in the future to track the development and progression of MAFLD. Supplementary Information The online version contains supplementary material available at 10.1186/s12944-022-01637-7.
Collapse
Affiliation(s)
- Qian Ren
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.,Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China.,Luzhou Key Laboratory of Cardiovascular and Metabolic Diseases, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.,Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
| | - Hongya Wang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.,Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China.,Luzhou Key Laboratory of Cardiovascular and Metabolic Diseases, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.,Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
| | - Yan Zeng
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.,Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China.,Luzhou Key Laboratory of Cardiovascular and Metabolic Diseases, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.,Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
| | - Xia Fang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.,Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China.,Luzhou Key Laboratory of Cardiovascular and Metabolic Diseases, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.,Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, China.,Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Mei Wang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.,Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China.,Luzhou Key Laboratory of Cardiovascular and Metabolic Diseases, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.,Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
| | - Dongze Li
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.,Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China.,Luzhou Key Laboratory of Cardiovascular and Metabolic Diseases, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.,Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
| | - Wei Huang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China. .,Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China. .,Luzhou Key Laboratory of Cardiovascular and Metabolic Diseases, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China. .,Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, China.
| | - Yong Xu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China. .,Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China. .,Luzhou Key Laboratory of Cardiovascular and Metabolic Diseases, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China. .,Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, China.
| |
Collapse
|
114
|
Eriksen PL, Thomsen KL, Sørensen M, Vilstrup H, Hvas AM. Impaired fibrinolysis without hypercoagulability characterises patients with non-alcoholic fatty liver disease. Thromb Res 2022; 213:9-15. [DOI: 10.1016/j.thromres.2022.02.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 02/14/2022] [Accepted: 02/24/2022] [Indexed: 12/12/2022]
|
115
|
Maevskaya M, Kotovskaya Y, Ivashkin V, Tkacheva O, Troshina E, Shestakova M, Breder V, Geyvandova N, Doschitsin V, Dudinskaya E, Ershova E, Kodzoeva K, Komshilova K, Korochanskaya N, Mayorov A, Mishina E, Nadinskaya M, Nikitin I, Pogosova N, Tarzimanova A, Shamkhalova M. The National Consensus statement on the management of adult patients with non-alcoholic fatty liver disease and main comorbidities. TERAPEVT ARKH 2022; 94:216-253. [DOI: 10.26442/00403660.2022.02.201363] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Indexed: 12/15/2022]
Abstract
The National Consensus was prepared with the participation of the National Medical Association for the Study of the Multimorbidity, Russian Scientific Liver Society, Russian Association of Endocrinologists, Russian Association of Gerontologists and Geriatricians, National Society for Preventive Cardiology, Professional Foundation for the Promotion of Medicine Fund PROFMEDFORUM.
The aim of the multidisciplinary consensus is a detailed analysis of the course of non-alcoholic fatty liver disease (NAFLD) and the main associated conditions. The definition of NAFLD is given, its prevalence is described, methods for diagnosing its components such as steatosis, inflammation and fibrosis are described.
The association of NAFLD with a number of cardio-metabolic diseases (arterial hypertension, atherosclerosis, thrombotic complications, type 2 diabetes mellitus (T2DM), obesity, dyslipidemia, etc.), chronic kidney disease (CKD) and the risk of developing hepatocellular cancer (HCC) were analyzed. The review of non-drug methods of treatment of NAFLD and modern opportunities of pharmacotherapy are presented.
The possibilities of new molecules in the treatment of NAFLD are considered: agonists of nuclear receptors, antagonists of pro-inflammatory molecules, etc. The positive properties and disadvantages of currently used drugs (vitamin E, thiazolidinediones, etc.) are described. Special attention is paid to the multi-target ursodeoxycholic acid (UDCA) molecule in the complex treatment of NAFLD as a multifactorial disease. Its anti-inflammatory, anti-oxidant and cytoprotective properties, the ability to reduce steatosis an independent risk factor for the development of cardiovascular pathology, reduce inflammation and hepatic fibrosis through the modulation of autophagy are considered.
The ability of UDCA to influence glucose and lipid homeostasis and to have an anticarcinogenic effect has been demonstrated. The Consensus statement has advanced provisions for practitioners to optimize the diagnosis and treatment of NAFLD and related common pathogenetic links of cardio-metabolic diseases.
Collapse
|
116
|
Cabrera D, Rao I, Raasch F, Solis N, Pizarro M, Freire M, Sáenz De Urturi D, Ramírez CA, Triantafilo N, León J, Riquelme A, Barrera F, Baudrand R, Aspichueta P, Arrese M, Arab JP. Mineralocorticoid receptor modulation by dietary sodium influences NAFLD development in mice. Ann Hepatol 2022; 24:100357. [PMID: 33940220 DOI: 10.1016/j.aohep.2021.100357] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/02/2021] [Accepted: 03/02/2021] [Indexed: 02/04/2023]
Abstract
INTRODUCTION AND OBJECTIVES Nonalcoholic-fatty-liver disease (NAFLD) is considered the hepatic manifestation of metabolic syndrome (MetS). Mineralocorticoid receptor (MR) activation is associated with increased risk of MetS but few studies have assessed the role of liver MR on NAFLD. We aimed to evaluate the effect of MR modulation by sodium intake in liver injury in experimental models of NAFLD. MATERIALS AND METHODS C57BL/6J mice were fed either a high-fat-diet (HFD) or a choline/methionine deficient (MCD) diet with different sodium concentrations. Hepatic concentration of lipid species, serum aldosterone levels, expression of MR, proinflammatory and profibrotic markers and liver histology were assessed. RESULTS Mice fed with High-Na+/HFD showed a lower MR expression in liver (p = 0.01) and less steatosis on histology (p = 0.04). Consistently, animals from this group exhibited lower levels of serum aldosterone (p = 0.028) and lower hepatic triglyceride content (p = 0.008). This associated to a reduced expression of lipogenic genes, significant changes in lipid subspecies, lower HOMA-IR (p < 0.05), and lower expression of pro-inflammatory and profibrotic markers compared to those mice fed a Low-Na+/HFD. Additionally, mice fed a High-Na+/HFD showed higher expression of salt-inducible kinase (SIK)-1 and lower expression of serum-and-glucocorticoid-inducible kinase (SGK)-1. Similar results were observed with the MCD diet model. CONCLUSION We identified in two experimental models of NAFLD that High-Na+ diet content is associated to lower serum aldosterone levels and hepatic MR downregulation, associated to decreased steatosis and reduced de novo hepatic lipogenesis, proinflammatory and profibrotic markers. Decreased activation of hepatic MR seems to generate beneficial downstream inhibition of lipogenesis in experimental NAFLD.
Collapse
Affiliation(s)
- Daniel Cabrera
- Departamento de Gastroenterologia, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile; Facultad de Ciencias Médicas, Universidad Bernardo O Higgins, Santiago, Chile
| | - Isabel Rao
- Departamento de Gastroenterologia, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Fabiola Raasch
- Departamento de Gastroenterologia, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Nancy Solis
- Departamento de Gastroenterologia, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Margarita Pizarro
- Departamento de Gastroenterologia, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Mariela Freire
- Departamento de Gastroenterologia, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | | | | | - Nicolás Triantafilo
- Departamento de Hematologia, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Jonathan León
- Departamento de Gastroenterologia, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Arnoldo Riquelme
- Departamento de Gastroenterologia, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile; Departamento de Ciencias de la Salud, Facultad de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Francisco Barrera
- Departamento de Gastroenterologia, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Rene Baudrand
- Departamento de Endocrinologia, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Patricia Aspichueta
- Department of Physiology, University of the Basque Country UPV/EHU, Leioa, Spain; Biocruces Health Research Institute, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Spain
| | - Marco Arrese
- Departamento de Gastroenterologia, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile; Centro de Envejecimiento y Regeneracion (CARE), Departamento de Biologia Celular y Molecular, Facultad de Ciencias Biologicas Pontificia Universidad Catolica de Chile, Santiago, Chile.
| | - Juan P Arab
- Departamento de Gastroenterologia, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile; Centro de Envejecimiento y Regeneracion (CARE), Departamento de Biologia Celular y Molecular, Facultad de Ciencias Biologicas Pontificia Universidad Catolica de Chile, Santiago, Chile.
| |
Collapse
|
117
|
Gao W, Xu B, Zhang Y, Liu S, Duan Z, Chen Y, Zhang X. Baicalin Attenuates Oxidative Stress in a Tissue-Engineered Liver Model of NAFLD by Scavenging Reactive Oxygen Species. Nutrients 2022; 14:541. [PMID: 35276900 PMCID: PMC8840060 DOI: 10.3390/nu14030541] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 12/14/2022] Open
Abstract
Oxidative stress plays an important role in the pathogenesis of nonalcoholic fatty liver disease (NAFLD). Baicalin has been shown to exert protective effects in various liver diseases. The mechanism of baicalin's antioxidative effect in NAFLD is currently unclear. The aim of this study was to investigate the effects and mechanisms of baicalin on oxidative stress in a new tissue-engineered liver model of NAFLD. The 3D model of NAFLD was induced by a fat-supplemented medium (fatty acids, FFA group) for 8 days and baicalin was administered on the 5th day. CCK-8 assay showed that baicalin at concentrations below 100 μM had no obvious cytotoxicity. Baicalin inhibited apoptosis and lactate dehydrogenase release in the FFA group. Baicalin reduced the levels of reactive oxygen species and malondialdehyde induced by FFA, and increased superoxide dismutase and glutathione amounts. However, it did not upregulate nuclear erythroid 2-related factor 2 compared with the FFA group. Mitochondrial morphology was partially restored after baicalin treatment, and ATP5A expression and mitochondrial membrane potential were increased. The superoxide anion scavenging ability of baicalin was enhanced in a dose-dependent manner. In summary, baicalin reduces oxidative stress and protects the mitochondria to inhibit apoptosis in the 3D NAFLD model via its own antioxidant activity.
Collapse
Affiliation(s)
- Wen Gao
- Department II of Liver Diseases, Beijing Youan Hospital Affiliated to Capital Medical University, Beijing 100069, China; (W.G.); (B.X.)
| | - Bin Xu
- Department II of Liver Diseases, Beijing Youan Hospital Affiliated to Capital Medical University, Beijing 100069, China; (W.G.); (B.X.)
| | - Yizhi Zhang
- Beijing Key Laboratory of Liver Failure and Artificial Liver Treatment, Department IV of Liver Diseases, Beijing Youan Hospital Affiliated to Capital Medical University, Beijing 100069, China; (Y.Z.); (S.L.); (Z.D.)
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing 100069, China
| | - Shuang Liu
- Beijing Key Laboratory of Liver Failure and Artificial Liver Treatment, Department IV of Liver Diseases, Beijing Youan Hospital Affiliated to Capital Medical University, Beijing 100069, China; (Y.Z.); (S.L.); (Z.D.)
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing 100069, China
| | - Zhongping Duan
- Beijing Key Laboratory of Liver Failure and Artificial Liver Treatment, Department IV of Liver Diseases, Beijing Youan Hospital Affiliated to Capital Medical University, Beijing 100069, China; (Y.Z.); (S.L.); (Z.D.)
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing 100069, China
| | - Yu Chen
- Beijing Key Laboratory of Liver Failure and Artificial Liver Treatment, Department IV of Liver Diseases, Beijing Youan Hospital Affiliated to Capital Medical University, Beijing 100069, China; (Y.Z.); (S.L.); (Z.D.)
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing 100069, China
| | - Xiaohui Zhang
- Beijing Key Laboratory of Liver Failure and Artificial Liver Treatment, Department IV of Liver Diseases, Beijing Youan Hospital Affiliated to Capital Medical University, Beijing 100069, China; (Y.Z.); (S.L.); (Z.D.)
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing 100069, China
| |
Collapse
|
118
|
Tomar M, Somvanshi PR, Kareenhalli V. Physiological significance of bistable circuit design in metabolic homeostasis: role of integrated insulin-glucagon signalling network. Mol Biol Rep 2022; 49:5017-5028. [DOI: 10.1007/s11033-022-07175-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 01/19/2022] [Indexed: 10/19/2022]
|
119
|
The Association between Serum Uric Acid Levels and 10-Year Cardiovascular Disease Risk in Non-Alcoholic Fatty Liver Disease Patients. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19031042. [PMID: 35162067 PMCID: PMC8834479 DOI: 10.3390/ijerph19031042] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/10/2022] [Accepted: 01/17/2022] [Indexed: 02/04/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) and serum uric acid (SUA) levels are risk factors for developing cardiovascular disease (CVD). Additionally, previous studies have suggested that high SUA levels increase the risk of having NAFLD. However, no study has investigated the relationship between SUA and CVD risk in NAFLD. This study analyzed the relationship between SUA and CVD in NAFLD. Data for this study used the 2016–2018 Korean National Health and Nutrition Examination Survey, which represents the Korean population. A total of 11,160 NAFLD patients were included. Participants with hepatic steatosis index ≥ 30 were considered to have NAFLD. Ten-year CVD risk was estimated using an integer-based Framingham risk score. Estimated 10-year CVD risk ≥ 20% was considered high risk. Multiple logistic regression was conducted to calculate the odds ratios (ORs) associated with SUA level and CVD risk. High CVD risk OR increases by 1.31 (95% CI 1.26–1.37) times per 1 mg/dL of SUA. After adjustment, SUA still had an increased risk (OR 1.44; 95% CI 1.38–1.51) of CVD. Compared with the lowest SUA quartile group, the highest quartile group showed a significantly higher risk of having CVD before (OR 2.76; 95% CI 2.34–3.25) and after (OR 4.01; 95% CI 3.37–4.78) adjustment. SUA is independently associated with CVS risk in NAFLD.
Collapse
|
120
|
Lampignano L, Donghia R, Sila A, Bortone I, Tatoli R, De Nucci S, Castellana F, Zupo R, Tirelli S, Giannoccaro V, Guerra V, Panza F, Lozupone M, Mastronardi M, De Pergola G, Giannelli G, Sardone R. Mediterranean Diet and Fatty Liver Risk in a Population of Overweight Older Italians: A Propensity Score-Matched Case-Cohort Study. Nutrients 2022; 14:nu14020258. [PMID: 35057439 PMCID: PMC8779579 DOI: 10.3390/nu14020258] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 12/31/2021] [Accepted: 01/05/2022] [Indexed: 12/21/2022] Open
Abstract
Hepatic steatosis, often known as fatty liver, is the most common hepatic disease in Western countries. The latest guidelines for the treatment of nonalcoholic fatty liver disease emphasize lifestyle measures, such as changing unhealthy eating patterns. Using a propensity score-matching approach, this study investigated the effect of adhering to a Mediterranean diet (MedDiet) on fatty liver risk in an older population (≥65 years) from Southern Italy. We recruited 1.403 subjects (53.6% men, ≥65 years) who completed a food frequency questionnaire (FFQ) and underwent clinical assessment between 2015 and 2018. For the assessment of the liver fat content, we applied the Fatty Liver Index (FLI). To evaluate the treatment effect of the MedDiet, propensity score matching was performed on patients with and without FLI > 60. After propensity score-matching with the MedDiet pattern as treatment, we found a higher consumption of red meat (p = 0.04) and wine (p = 0.04) in subjects with FLI > 60. Based on the FLI, the inverse association shown between adherence to the MedDiet and the risk of hepatic steatosis shows that the MedDiet can help to prevent hepatic steatosis. Consuming less red and processed meat, as well as alcoholic beverages, may be part of these healthy lifestyle recommendations.
Collapse
Affiliation(s)
- Luisa Lampignano
- Unit of Data Sciences and Technology Innovation for Population Health, National Institute of Gastroenterology “Saverio de Bellis”, Research Hospital, 70013 Bari, Italy; (L.L.); (R.D.); (A.S.); (I.B.); (R.T.); (S.D.N.); (F.C.); (R.Z.); (S.T.); (V.G.)
| | - Rossella Donghia
- Unit of Data Sciences and Technology Innovation for Population Health, National Institute of Gastroenterology “Saverio de Bellis”, Research Hospital, 70013 Bari, Italy; (L.L.); (R.D.); (A.S.); (I.B.); (R.T.); (S.D.N.); (F.C.); (R.Z.); (S.T.); (V.G.)
| | - Annamaria Sila
- Unit of Data Sciences and Technology Innovation for Population Health, National Institute of Gastroenterology “Saverio de Bellis”, Research Hospital, 70013 Bari, Italy; (L.L.); (R.D.); (A.S.); (I.B.); (R.T.); (S.D.N.); (F.C.); (R.Z.); (S.T.); (V.G.)
| | - Ilaria Bortone
- Unit of Data Sciences and Technology Innovation for Population Health, National Institute of Gastroenterology “Saverio de Bellis”, Research Hospital, 70013 Bari, Italy; (L.L.); (R.D.); (A.S.); (I.B.); (R.T.); (S.D.N.); (F.C.); (R.Z.); (S.T.); (V.G.)
| | - Rossella Tatoli
- Unit of Data Sciences and Technology Innovation for Population Health, National Institute of Gastroenterology “Saverio de Bellis”, Research Hospital, 70013 Bari, Italy; (L.L.); (R.D.); (A.S.); (I.B.); (R.T.); (S.D.N.); (F.C.); (R.Z.); (S.T.); (V.G.)
| | - Sara De Nucci
- Unit of Data Sciences and Technology Innovation for Population Health, National Institute of Gastroenterology “Saverio de Bellis”, Research Hospital, 70013 Bari, Italy; (L.L.); (R.D.); (A.S.); (I.B.); (R.T.); (S.D.N.); (F.C.); (R.Z.); (S.T.); (V.G.)
| | - Fabio Castellana
- Unit of Data Sciences and Technology Innovation for Population Health, National Institute of Gastroenterology “Saverio de Bellis”, Research Hospital, 70013 Bari, Italy; (L.L.); (R.D.); (A.S.); (I.B.); (R.T.); (S.D.N.); (F.C.); (R.Z.); (S.T.); (V.G.)
| | - Roberta Zupo
- Unit of Data Sciences and Technology Innovation for Population Health, National Institute of Gastroenterology “Saverio de Bellis”, Research Hospital, 70013 Bari, Italy; (L.L.); (R.D.); (A.S.); (I.B.); (R.T.); (S.D.N.); (F.C.); (R.Z.); (S.T.); (V.G.)
| | - Sarah Tirelli
- Unit of Data Sciences and Technology Innovation for Population Health, National Institute of Gastroenterology “Saverio de Bellis”, Research Hospital, 70013 Bari, Italy; (L.L.); (R.D.); (A.S.); (I.B.); (R.T.); (S.D.N.); (F.C.); (R.Z.); (S.T.); (V.G.)
| | | | - Vito Guerra
- Unit of Data Sciences and Technology Innovation for Population Health, National Institute of Gastroenterology “Saverio de Bellis”, Research Hospital, 70013 Bari, Italy; (L.L.); (R.D.); (A.S.); (I.B.); (R.T.); (S.D.N.); (F.C.); (R.Z.); (S.T.); (V.G.)
| | - Francesco Panza
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, 11, 70125 Bari, Italy; (F.P.); (M.L.)
| | - Madia Lozupone
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, 11, 70125 Bari, Italy; (F.P.); (M.L.)
| | - Mauro Mastronardi
- Inflammatory Bowel Disease Unit, National Institute of Gastroenterology, “Saverio De Bellis” Research Hospital, 70013 Bari, Italy;
| | - Giovanni De Pergola
- Unit of Geriatrics and Internal Medicine, National Institute of Gastroenterology “Saverio de Bellis”, Research Hospital, 70013 Bari, Italy;
| | - Gianluigi Giannelli
- Scientific Direction, National Institute of Gastroenterology, “Saverio De Bellis” Research Hospital, 70013 Bari, Italy;
| | - Rodolfo Sardone
- Unit of Data Sciences and Technology Innovation for Population Health, National Institute of Gastroenterology “Saverio de Bellis”, Research Hospital, 70013 Bari, Italy; (L.L.); (R.D.); (A.S.); (I.B.); (R.T.); (S.D.N.); (F.C.); (R.Z.); (S.T.); (V.G.)
- Correspondence:
| |
Collapse
|
121
|
Kim H, Zhang D, Song Z, Tong X, Zhang K. Analysis of Insulin Resistance in Nonalcoholic Steatohepatitis. Methods Mol Biol 2022; 2455:233-241. [PMID: 35212998 PMCID: PMC9053411 DOI: 10.1007/978-1-0716-2128-8_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Insulin resistance is a major phenotype observed in nonalcoholic steatohepatitis (NASH), the advanced stage of nonalcoholic fatty liver disease (NAFLD). Insulin resistance in NASH is characterized by reductions in whole body, hepatic, and adipose tissue insulin sensitivity. The mechanisms underlying hepatic insulin resistance is primarily associated with hepatic glucose production (HGP) rate. Hepatic insulin resistance can also be a consequence or a driving factor of hepatic lipid accumulation by increasing free fatty acid synthesis, delivery, and catabolism. The common method to assess hepatic insulin resistance is to measure hepatic glucose production (HGP) using isotope tracer distribution technique. However, non-radioactive approaches have been developed to assess hepatic insulin resistance in the context of NASH. In this chapter, we describe the methods to evaluate hepatic insulin resistance in animal models of NASH by examining insulin sensitivity and glucose tolerance as well as the key molecules in hepatic insulin signaling pathways.
Collapse
Affiliation(s)
- Hyunbae Kim
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Deqiang Zhang
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Zhenfeng Song
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Xin Tong
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA.
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI, USA.
| |
Collapse
|
122
|
Jiang LP, Sun HZ. Long-chain saturated fatty acids and its interaction with insulin resistance and the risk of nonalcoholic fatty liver disease in type 2 diabetes in Chinese. Front Endocrinol (Lausanne) 2022; 13:1051807. [PMID: 36568120 PMCID: PMC9768420 DOI: 10.3389/fendo.2022.1051807] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/23/2022] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION This study aimed to explore relationships between long-chain saturated fatty acids (LSFAs) and nonalcoholic fatty liver disease (NAFLD) in patients with type 2 diabetes (T2D); and whether insulin action had an interactive effect with LSFAs on NAFLD progression. METHODS From April 2018 to April 2019, we extracted the electronic medical records of 481 patients with T2D who meet the inclusion and exclusion criteria from the Second Affiliated Hospital of Dalian Medical University. Ultrasound was used to estimate NAFLD at admission. Logistic regression analysis were used to estimate odds ratios (OR) and 95% confidence intervals (CI). The additive interaction was carried out to estimate interactions between LSFAs and insulin resistance (IR) in NAFLD patients with T2D. RESULTS Myristic acid (14:0) and palmitic acid (16:0) were positively associated with the risk of NAFLD (OR for myristic acid (14:0): 7.516, 3.557-15.882 and OR for palmitic acid (16:0): 4.071, 1.987-8.343, respectively). After adjustment for traditional risk factors, these associations were slightly attenuated but still highly significant. Co-presence of myristic acid (14:0)>72.83 μmol/L and IR>4.89 greatly increased OR of NAFLD to 9.691 (4.113-22.833). Similarly, co-presence of palmitic acid (16:0)>3745.43μmol/L and IR>4.89 greatly increased OR of NAFLD to 6.518(2.860-14.854). However, stearic acid (18:0) and risk of NAFLD have no association. Moreover, there was no association between very-long-chain SFAs (VLSFAs) and risk of NAFLD. DISCUSSION Myristic acid (14:0) and palmitic acid (16:0) were positively associated with the risk of NAFLD in T2D patients in China. High IR amplified the effect of high myristic acid (14:0) and high palmitic acid (16:0) on NAFLD.
Collapse
Affiliation(s)
- Li-Peng Jiang
- Department of Radiation Oncology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Hong-Zhi Sun
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
- Key Laboratory of Liaoning Tumor Clinical Metabolomics (KLLTCM), Jinzhou Medical University, Jinzhou, China
- *Correspondence: Hong-Zhi Sun,
| |
Collapse
|
123
|
Qi L, Zushin PJ, Chang CF, Lee YT, Alba DL, Koliwad S, Stahl A. Probing Insulin Sensitivity with Metabolically Competent Human Stem Cell-Derived White Adipose Tissue Microphysiological Systems. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2103157. [PMID: 34761526 PMCID: PMC8776615 DOI: 10.1002/smll.202103157] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/21/2021] [Indexed: 05/13/2023]
Abstract
Impaired white adipose tissue (WAT) function has been recognized as a critical early event in obesity-driven disorders, but high buoyancy, fragility, and heterogeneity of primary adipocytes have largely prevented their use in drug discovery efforts highlighting the need for human stem cell-based approaches. Here, human stem cells are utilized to derive metabolically functional 3D adipose tissue (iADIPO) in a microphysiological system (MPS). Surprisingly, previously reported WAT differentiation approaches create insulin resistant WAT ill-suited for type-2 diabetes mellitus drug discovery. Using three independent insulin sensitivity assays, i.e., glucose and fatty acid uptake and suppression of lipolysis, as the functional readouts new differentiation conditions yielding hormonally responsive iADIPO are derived. Through concomitant optimization of an iADIPO-MPS, it is abled to obtain WAT with more unilocular and significantly larger (≈40%) lipid droplets compared to iADIPO in 2D culture, increased insulin responsiveness of glucose uptake (≈2-3 fold), fatty acid uptake (≈3-6 fold), and ≈40% suppressing of stimulated lipolysis giving a dynamic range that is competent to current in vivo and ex vivo models, allowing to identify both insulin sensitizers and desensitizers.
Collapse
Affiliation(s)
- Lin Qi
- Department of Nutritional Science and Toxicology, College of Natural Resources, University of California, Berkeley, Berkeley, California, 94720, USA
| | - Peter James Zushin
- Department of Nutritional Science and Toxicology, College of Natural Resources, University of California, Berkeley, Berkeley, California, 94720, USA
| | - Ching-Fang Chang
- Department of Nutritional Science and Toxicology, College of Natural Resources, University of California, Berkeley, Berkeley, California, 94720, USA
| | - Yue Tung Lee
- Department of Nutritional Science and Toxicology, College of Natural Resources, University of California, Berkeley, Berkeley, California, 94720, USA
| | - Diana L. Alba
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of California, San Francisco; Diabetes Center, University of California, San Francisco, San Francisco, California 94143, USA
| | - Suneil Koliwad
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of California, San Francisco; Diabetes Center, University of California, San Francisco, San Francisco, California 94143, USA
| | - Andreas Stahl
- Department of Nutritional Science and Toxicology, College of Natural Resources, University of California, Berkeley, Berkeley, California, 94720, USA
| |
Collapse
|
124
|
Yin H, Shi A, Wu J. Platelet-Activating Factor Promotes the Development of Non-Alcoholic Fatty Liver Disease. Diabetes Metab Syndr Obes 2022; 15:2003-2030. [PMID: 35837578 PMCID: PMC9275506 DOI: 10.2147/dmso.s367483] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/28/2022] [Indexed: 11/23/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a multifaceted clinicopathological syndrome characterised by excessive hepatic lipid accumulation that causes steatosis, excluding alcoholic factors. Platelet-activating factor (PAF), a biologically active lipid transmitter, induces platelet activation upon binding to the PAF receptor. Recent studies have found that PAF is associated with gamma-glutamyl transferase, which is an indicator of liver disease. Moreover, PAF can stimulate hepatic lipid synthesis and cause hypertriglyceridaemia. Furthermore, the knockdown of the PAF receptor gene in the animal models of NAFLD helped reduce the inflammatory response, improve glucose homeostasis and delay the development of NAFLD. These findings suggest that PAF is associated with NAFLD development. According to reports, patients with NAFLD or animal models have marked platelet activation abnormalities, mainly manifested as enhanced platelet adhesion and aggregation and altered blood rheology. Pharmacological interventions were accompanied by remission of abnormal platelet activation and significant improvement in liver function and lipids in the animal model of NAFLD. These confirm that platelet activation may accompany a critical importance in NAFLD development and progression. However, how PAFs are involved in the NAFLD signalling pathway needs further investigation. In this paper, we review the relevant literature in recent years and discuss the role played by PAF in NAFLD development. It is important to elucidate the pathogenesis of NAFLD and to find effective interventions for treatment.
Collapse
Affiliation(s)
- Hang Yin
- Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, People’s Republic of China
| | - Anhua Shi
- Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, People’s Republic of China
| | - Junzi Wu
- Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, People’s Republic of China
- Correspondence: Junzi Wu; Anhua Shi, Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, People’s Republic of China, Tel/Fax +86 187 8855 7524; +86 138 8885 0813, Email ;
| |
Collapse
|
125
|
El Jamaly H, Eslick GD, Weltman M. Systematic review with meta-analysis: Non-alcoholic fatty liver disease and the association with pregnancy outcomes. Clin Mol Hepatol 2022; 28:52-66. [PMID: 34530527 PMCID: PMC8755467 DOI: 10.3350/cmh.2021.0205] [Citation(s) in RCA: 8] [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: 07/16/2021] [Revised: 09/13/2021] [Accepted: 09/16/2021] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND/AIMS Maternal and fetal outcomes in pregnant patients with Non-alcoholic fatty liver disease (NAFLD) have been largely unexplored. To determine the level of evidence associated with maternal and fetal outcomes in pregnant women with NAFLD. METHODS We conducted a comprehensive literature search. The studies included pregnant patients with a previous, current or subsequent diagnosis of NAFLD. We used a random-effects model using odds ratios (OR) with 95% confidence intervals (CI). RESULTS Twenty-two studies, with 13,641 female NAFLD patients were reviewed. The results highlight that NAFLD patients had a statistically significant increased likelihood of baseline diabetes mellitus (OR, 6.00; 95% CI, 2.21-16.31; P<0.001; n=7), baseline Hypertension (OR, 3.75; 95% CI, 2.13-6.59; P<0.001; n=4), gestational hypertension (OR, 1.83; 95% CI, 1.03-3.26; P=0.041; n=2), and pre-eclampsia (OR, 2.43; 95% CI, 1.46-4.04; P=0.001; n=3). The odds for a past and current history of gestational diabetes mellitus were OR, 3.78; 95% CI, 2.21-6.44; P<0.001; n=5 and OR, 3.23; 95% CI, 1.97- 5.31; P<0.001; n=6, respectively. As for fetal outcomes, pregnant NAFLD patients were significantly more likely to have a premature birth (OR, 2.02; 95% CI, 1.44-2.85; P<0.001; n=4), large for gestational age birth (OR, 2.01; 95% CI, 1.72-2.37; P<0.001; n=2) or a history of prior miscarriage or abortion (OR, 1.15; 95% CI, 1.02-1.30; P=0.02; n=2). Egger's regression revealed no evidence of publication bias (P>0.05). CONCLUSION This meta-analysis provides pooled evidence that NAFLD is associated with a substantial increase in maternal diabetic and hypertensive complications and multiple adverse fetal outcomes. This data is important for clinicians managing these patients before, during and after pregnancy.
Collapse
Affiliation(s)
- Hydar El Jamaly
- Department of Gastroenterology and Hepatology, Nepean Hospital, Penrith, Australia
- Nepean Clinical School, The University of Sydney, Penrith, Australia
| | - Guy D Eslick
- The Centre for Digestive Health and Neurogastroenterology, Hunter Medical Research Institute, The University of Newcastle, Newcastle, Australia
| | - Martin Weltman
- Department of Gastroenterology and Hepatology, Nepean Hospital, Penrith, Australia
- Nepean Clinical School, The University of Sydney, Penrith, Australia
| |
Collapse
|
126
|
Luo Y, Lu H, Peng D, Ruan X, Chen YE, Guo Y. Liver-humanized mice: A translational strategy to study metabolic disorders. J Cell Physiol 2022; 237:489-506. [PMID: 34661916 PMCID: PMC9126562 DOI: 10.1002/jcp.30610] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/07/2021] [Accepted: 09/11/2021] [Indexed: 01/03/2023]
Abstract
The liver is the metabolic core of the whole body. Tools commonly used to study the human liver metabolism include hepatocyte cell lines, primary human hepatocytes, and pluripotent stem cells-derived hepatocytes in vitro, and liver genetically humanized mouse model in vivo. However, none of these systems can mimic the human liver in physiological and pathological states satisfactorily. Liver-humanized mice, which are established by reconstituting mouse liver with human hepatocytes, have emerged as an attractive animal model to study drug metabolism and evaluate the therapeutic effect in "human liver" in vivo because the humanized livers greatly replicate enzymatic features of human hepatocytes. The application of liver-humanized mice in studying metabolic disorders is relatively less common due to the largely uncertain replication of metabolic profiles compared to humans. Here, we summarize the metabolic characteristics and current application of liver-humanized mouse models in metabolic disorders that have been reported in the literature, trying to evaluate the pros and cons of using liver-humanized mice as novel mouse models to study metabolic disorders.
Collapse
Affiliation(s)
- Yonghong Luo
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Haocheng Lu
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Daoquan Peng
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Xiangbo Ruan
- Division of Endocrinology, Diabetes and Metabolism, Johns Hopkins School of Medicine, Johns Hopkins All Children’s Hospital, St. Petersburg, FL 33701, USA
| | - Y. Eugene Chen
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
- Center for Advanced Models and Translational Sciences and Therapeutics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yanhong Guo
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| |
Collapse
|
127
|
Abnormal transaminase and lipid profiles in coexisting diseases in patients with fatty liver: a population study in Sichuan. Biosci Rep 2021; 41:230168. [PMID: 34918746 PMCID: PMC8685641 DOI: 10.1042/bsr20211769] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 11/02/2021] [Accepted: 11/03/2021] [Indexed: 02/05/2023] Open
Abstract
Among chronic liver diseases, fatty liver has the highest incidence worldwide. Coexistence of fatty liver and other chronic diseases, such as diabetes, hepatitis B virus (HBV) and Helicobacter pylori (Hp) infection, is common in clinical practice. The present study was conducted to analyze the prevalence and association of coexisting diseases in patients with fatty liver and to investigate how coexisting diseases contribute to abnormal transaminase and lipid profiles. We enrolled participants who were diagnosed with fatty liver via ultrasound in the physical examination center of West China Hospital. Multivariable logistic regression was used to determine the adjusted odds ratios (ORs). We found that 23.6% of patients who underwent physical examinations were diagnosed with fatty liver. These patients had higher risks of metabolic syndrome (MetS), type 2 diabetes mellitus (T2DM), and hypertension and a lower risk of HBV infection. The risks of Hp infection and hyperthyroidism did not statistically differ. When fatty liver coexisted with T2DM, MetS and thyroid dysfunction, it conferred a higher risk of elevated transaminase. Fatty liver was positively correlated with triglycerides, cholesterol and low-density lipoprotein cholesterol (LDL-C) and negatively correlated with HBV; thus, HBV had a neutralizing effect on lipid metabolism when coexisting with fatty liver. In conclusion, patients with fatty liver that coexists with T2DM, MetS and thyroid dysfunction are more prone to elevated transaminase levels. Patients with both fatty liver and HBV may experience a neutralizing effect on their lipid metabolism. Thus, lipid alterations should be monitored in these patients during antiviral treatment for HBV.
Collapse
|
128
|
Role and Treatment of Insulin Resistance in Patients with Chronic Kidney Disease: A Review. Nutrients 2021; 13:nu13124349. [PMID: 34959901 PMCID: PMC8707041 DOI: 10.3390/nu13124349] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/26/2021] [Accepted: 11/30/2021] [Indexed: 01/11/2023] Open
Abstract
Patients with chronic kidney disease (CKD) and dialysis have higher mortality than those without, and cardiovascular disease (CVD) is the main cause of death. As CVD is caused by several mechanisms, insulin resistance plays an important role in CVD. This review summarizes the importance and mechanism of insulin resistance in CKD and discusses the current evidence regarding insulin resistance in patients with CKD and dialysis. Insulin resistance has been reported to influence endothelial dysfunction, plaque formation, hypertension, and dyslipidemia. A recent study also reported an association between insulin resistance and cognitive dysfunction, non-alcoholic fatty liver disease, polycystic ovary syndrome, and malignancy. Insulin resistance increases as renal function decrease in patients with CKD and dialysis. Several mechanisms increase insulin resistance in patients with CKD, such as chronic inflammation, oxidative stress, obesity, and mineral bone disorder. There is the possibility that insulin resistance is the potential future target of treatment in patients with CKD.
Collapse
|
129
|
Lu L, Chen C, Li Y, Guo W, Zhang S, Brockman J, Shikany JM, Kahe K. Magnesium intake is inversely associated with risk of non-alcoholic fatty liver disease among American adults. Eur J Nutr 2021; 61:1245-1254. [PMID: 34741649 DOI: 10.1007/s00394-021-02732-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 10/29/2021] [Indexed: 11/24/2022]
Abstract
PURPOSE Human data are limited linking magnesium (Mg) intake to the risk of non-alcoholic fatty liver disease (NAFLD). We aimed to examine the association between Mg intake and the risk of NAFLD among young adults in the US with a 25-year follow-up. METHODS This study included 2685 participants from the Coronary Artery Risk Development in Young Adult (CARDIA) study. Diet and dietary supplements were assessed at baseline (1985-1986) and exam years 7 and 20 using an interview-based dietary history. NAFLD, defined as liver attenuation ≤ 51 Hounsfield Units excluding secondary causes of liver fat accumulation, was identified by non-contrast-computed tomography scanning at exam year 25. Multivariable-adjusted logistic regression model was used to examine the associations between cumulative average total intake of Mg (dietary plus supplemental) and NAFLD odds. RESULTS A total of 629 NAFLD cases were documented. After adjustment for potential confounders, an inverse association between total Mg intake and NAFLD odds was observed. Compared to participants in the lowest quintile of total Mg intake, the odds of NAFLD was 55% lower among individuals in the highest quintile [multivariable-adjusted odds ratio (OR) = 0.45, 95% confidence interval (CI) (0.23, 0.85), p for trend = 0.03]. Consistently, whole-grain consumption, a major dietary source of Mg, was inversely associated with NAFLD odds (p for trend = 0.02). CONCLUSIONS This study suggests that higher cumulative intake of Mg throughout adulthood is associated with lower odds of NAFLD in midlife. Future studies are needed to establish a possible causal relationship.
Collapse
Affiliation(s)
- Liping Lu
- Department of Obstetrics and Gynecology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Cheng Chen
- Department of Obstetrics and Gynecology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Yuexia Li
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wenzhi Guo
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shuijun Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - John Brockman
- Department of Chemistry, University of Missouri, Columbia, MO, USA
| | - James M Shikany
- Division of Preventive Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ka Kahe
- Department of Obstetrics and Gynecology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA.
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA.
| |
Collapse
|
130
|
Li Y, Zhang S, Zhu Z, Zhou R, Xu P, Zhou L, Kan Y, Li J, Zhao J, Fang P, Yu X, Shang W. Upregulation of adiponectin by Ginsenoside Rb1 contributes to amelioration of hepatic steatosis induced by high fat diet. J Ginseng Res 2021; 46:561-571. [PMID: 35818425 PMCID: PMC9270646 DOI: 10.1016/j.jgr.2021.10.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 02/04/2023] Open
Abstract
Background Ginsenoside Rb1 (GRb1) is capable of regulating lipid and glucose metabolism through its action on adipocytes. However, the beneficial role of GRb1-induced up-regulation of adiponectin in liver steatosis remains unelucidated. Thus, we tested whether GRb1 ameliorates liver steatosis and insulin resistance by promoting the expression of adiponectin. Methods 3T3-L1 adipocytes and hepatocytes were used to investigate GRb1's action on adiponectin expression and triglyceride (TG) accumulation. Wild type (WT) mice and adiponectin knockout (KO) mice fed high fat diet were treated with GRb1 for 2 weeks. Hepatic fat accumulation and function as well as insulin sensitivity was measured. The activation of AMPK was also detected in the liver and hepatocytes. Results GRb1 reversed the reduction of adiponectin secretion in adipocytes. The conditioned medium (CM) from adipocytes treated with GRb1 reduced TG accumulation in hepatocytes, which was partly attenuated by the adiponectin antibody. In the KO mice, the GRb1-induced significant decrease of TG content, ALT and AST was blocked by the deletion of adiponectin. The elevations of GRb1-induced insulin sensitivity indicated by OGTT, ITT and HOMA-IR were also weakened in the KO mice. The CM treatment significantly enhanced the phosphorylation of AMPK in hepatocytes, but not GRb1 treatment. Likewise, the phosphorylation of AMPK in liver of the WT mice was increased by GRb1, but not in the KO mice. Conclusions The up-regulation of adiponectin by GRb1 contributes to the amelioration of liver steatosis and insulin resistance, which further elucidates a new mechanism underlying the beneficial effects of GRb1 on obesity.
Collapse
Affiliation(s)
- Yaru Li
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shuchen Zhang
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ziwei Zhu
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ruonan Zhou
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Pingyuan Xu
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lingyan Zhou
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yue Kan
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiao Li
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Juan Zhao
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Penghua Fang
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xizhong Yu
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Corresponding author.
| | - Wenbin Shang
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Corresponding author. Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
131
|
Goldberg RB, Tripputi MT, Boyko EJ, Budoff M, Chen ZZ, Clark JM, Dabelea DM, Edelstein SL, Gerszten RE, Horton E, Mather KJ, Perreault L, Temprosa M, Wallia A, Watson K, Irfan Z. Hepatic Fat in Participants With and Without Incident Diabetes in the Diabetes Prevention Program Outcome Study. J Clin Endocrinol Metab 2021; 106:e4746-e4765. [PMID: 33705543 PMCID: PMC8530730 DOI: 10.1210/clinem/dgab160] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Indexed: 11/19/2022]
Abstract
CONTEXT There is little information about fatty liver in prediabetes as it transitions to early diabetes. OBJECTIVE This study is aimed at evaluating the prevalence and determinants of fatty liver in the Diabetes Prevention Program (DPP). METHODS We measured liver fat as liver attenuation (LA) in Hounsfield units (HU) in 1876 participants at ~14 years following randomization into the DPP, which tested the effects of lifestyle or metformin interventions versus standard care to prevent diabetes. LA was compared among intervention groups and in those with versus without diabetes, and associations with baseline and follow-up measurements of anthropometric and metabolic covariates were assessed. RESULTS There were no differences in liver fat between treatment groups at 14 years of follow-up. Participants with diabetes had lower LA (mean ± SD: 46 ± 16 vs 51 ± 14 HU; P < 0.001) and a greater prevalence of fatty liver (LA < 40 HU) (34% vs 17%; P < 0.001). Severity of metabolic abnormalities at the time of LA evaluation was associated with lower LA categories in a graded manner and more strongly in those with diabetes. Averaged annual fasting insulin (an index of insulin resistance [OR, 95% CI 1.76, 1.41-2.20]) waist circumference (1.63, 1.17-2.26), and triglyceride (1.42, 1.13-1.78), but not glucose, were independently associated with LA < 40 HU prevalence. CONCLUSION Fatty liver is common in the early phases of diabetes development. The association of LA with insulin resistance, waist circumference, and triglyceride levels emphasizes the importance of these markers for hepatic steatosis in this population and that assessment of hepatic fat in early diabetes development is warranted.
Collapse
Affiliation(s)
- Ronald B Goldberg
- Diabetes Research Institute, Division of Endocrinology, Diabetes and Metabolism, University of Miami Miller School of Medicine, Miami, FL 33136USA
- Correspondence: Ronald B Goldberg, MD, Diabetes Research Institute, Division of Endocrinology, Diabetes and Metabolism, University of Miami Miller School of Medicine, 1450 Northwest 10th Avenue, Miami, FL 33136 USA.
| | - Mark T Tripputi
- Milken Institute School of Public Health and Health Services, The Biostatistics Center, George Washington University, Rockville, MD 20852USA
| | - Edward J Boyko
- University of Washington, Department of Medicine, Seattle, WA 98108USA
| | - Matthew Budoff
- Los Angeles Biomedical Research Institute, Torrance CA 90502USA
| | - Zsu-Zsu Chen
- Division of Endocrinology, Bone, and Metabolism, Beth Israel Deaconess Medical Center, Harvard, Boston, MA 02215, USA
| | - Jeanne M Clark
- Division of General Internal Medicine, The Johns Hopkins University, Baltimore MD 21287USA
| | - Dana M Dabelea
- Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO 80045USA
| | - Sharon L Edelstein
- Milken Institute School of Public Health and Health Services, The Biostatistics Center, George Washington University, Rockville, MD 20852USA
- Sharon L. Edelstein, ScM, The Biostatistics Center, Milken Institute School of Public Health, The George Washington University, 6110 Executive Blvd., Suite 750, Rockville, MD 20852, USA.
| | - Robert E Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard, Boston, MA 02215, USA
| | | | | | - Leigh Perreault
- University of Colorado Anschutz Medical Campus, Aurora, CO 80045USA
| | - Marinella Temprosa
- Milken Institute School of Public Health and Health Services, The Biostatistics Center, George Washington University, Rockville, MD 20852USA
| | - Amisha Wallia
- Northwestern University Feinberg School of Medicine, Chicago IL 60610USA
| | - Karol Watson
- David Geffen School of Medicine at UCLA, Los Angeles, CA 90095USA
| | - Zeb Irfan
- J. W. Ruby Memorial Hospital, Morgantown, WV 26505USA
| |
Collapse
|
132
|
Zou Y, Lan J, Zhong Y, Yang S, Zhang H, Xie G. Association of remnant cholesterol with nonalcoholic fatty liver disease: a general population-based study. Lipids Health Dis 2021; 20:139. [PMID: 34657611 PMCID: PMC8520640 DOI: 10.1186/s12944-021-01573-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 10/05/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Remnant cholesterol (RC) mediates the progression of coronary artery disease, diabetic complications, hypertension, and chronic kidney disease. Limited information is available on the association of RC with nonalcoholic fatty liver disease (NAFLD). This study aimed to explore whether RC can be used to independently evaluate the risk of NAFLD in the general population and to analyze the predictive value of RC for NAFLD. METHODS The study included 14,251 subjects enrolled in a health screening program. NAFLD was diagnosed by ultrasound, and the association of RC with NAFLD was assessed using the receiver operating characteristic (ROC) curve and logistic regression equation. RESULTS Subjects with elevated RC had a significantly higher risk of developing NAFLD after fully adjusting for potential confounding factors (OR 1.77 per SD increase, 95% CI 1.64-1.91, P trend< 0.001). There were significant differences in this association among sex, BMI and age stratification. Compared with men, women were facing a higher risk of RC-related NAFLD. Compared with people with normal BMI, overweight and obesity, the risk of RC-related NAFLD was higher in thin people. In different age stratifications, when RC increased, young people had a higher risk of developing NAFLD than other age groups. Additionally, ROC analysis results showed that among all lipid parameters, the AUC of RC was the largest (women: 0.81; men: 0.74), and the best threshold for predicting NAFLD was 0.54 in women and 0.63 in men. CONCLUSIONS The results obtained from this study indicate that (1) in the general population, RC is independently associated with NAFLD but not with other risk factors. (2) Compared with traditional lipid parameters, RC has a better predictive ability for NAFLD in men.
Collapse
Affiliation(s)
- Yang Zou
- From the Jiangxi Cardiovascular Research Institute, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, 330006, China
| | - Jianyun Lan
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, People's Republic of China, 330006
| | - Yanjia Zhong
- Department of Endocrinology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, People's Republic of China, 330006
| | - Shuo Yang
- Department of Cardiology, Dean County People's Hospital, Jiujiang, People's Republic of China, 330400
| | - Huimin Zhang
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, People's Republic of China, 330006
| | - Guobo Xie
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, People's Republic of China, 330006.
| |
Collapse
|
133
|
Ben-Haroush Schyr R, Al-Kurd A, Moalem B, Permyakova A, Israeli H, Bardugo A, Arad Y, Hefetz L, Bergel M, Haran A, Azar S, Magenheim I, Tam J, Grinbaum R, Ben-Zvi D. Sleeve Gastrectomy Suppresses Hepatic Glucose Production and Increases Hepatic Insulin Clearance Independent of Weight Loss. Diabetes 2021; 70:2289-2298. [PMID: 34341005 PMCID: PMC8576500 DOI: 10.2337/db21-0251] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 07/28/2021] [Indexed: 11/13/2022]
Abstract
Bariatric operations induce weight loss, which is associated with an improvement in hepatic steatosis and a reduction in hepatic glucose production. It is not clear whether these outcomes are entirely due to weight loss, or whether the new anatomy imposed by the surgery contributes to the improvement in the metabolic function of the liver. We performed vertical sleeve gastrectomy (VSG) on obese mice provided with a high-fat high-sucrose diet and compared them to diet and weight-matched sham-operated mice (WMS). At 40 days after surgery, VSG-operated mice displayed less hepatic steatosis compared with WMS. By measuring the fasting glucose and insulin levels in the blood vessels feeding and draining the liver, we showed directly that hepatic glucose production was suppressed after VSG. Insulin levels were elevated in the portal vein, and hepatic insulin clearance was elevated in VSG-operated mice. The hepatic expression of genes associated with insulin clearance was upregulated. We repeated the experiment in lean mice and observed that portal insulin and glucagon are elevated, but only insulin clearance is increased in VSG-operated mice. In conclusion, direct measurement of glucose and insulin in the blood entering and leaving the liver shows that VSG affects glucose and insulin metabolism through mechanisms independent of weight loss and diet.
Collapse
Affiliation(s)
- Rachel Ben-Haroush Schyr
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hadassah Medical School-The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Abbas Al-Kurd
- Department of Surgery, Hadassah Medical Center-Mt. Scopus, Jerusalem, Israel
| | - Botros Moalem
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hadassah Medical School-The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Anna Permyakova
- Obesity and Metabolism Laboratory, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Hadar Israeli
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hadassah Medical School-The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Aya Bardugo
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hadassah Medical School-The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yhara Arad
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hadassah Medical School-The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Liron Hefetz
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hadassah Medical School-The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Michael Bergel
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hadassah Medical School-The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Arnon Haran
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hadassah Medical School-The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Shahar Azar
- Obesity and Metabolism Laboratory, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Itia Magenheim
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hadassah Medical School-The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Joseph Tam
- Obesity and Metabolism Laboratory, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ronit Grinbaum
- Department of Surgery, Hadassah Medical Center-Mt. Scopus, Jerusalem, Israel
| | - Danny Ben-Zvi
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hadassah Medical School-The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
134
|
Koh EH, Yoon JE, Ko MS, Leem J, Yun JY, Hong CH, Cho YK, Lee SE, Jang JE, Baek JY, Yoo HJ, Kim SJ, Sung CO, Lim JS, Jeong WI, Back SH, Baek IJ, Torres S, Solsona-Vilarrasa E, Conde de la Rosa L, Garcia-Ruiz C, Feldstein AE, Fernandez-Checa JC, Lee KU. Sphingomyelin synthase 1 mediates hepatocyte pyroptosis to trigger non-alcoholic steatohepatitis. Gut 2021; 70:1954-1964. [PMID: 33208407 PMCID: PMC8458090 DOI: 10.1136/gutjnl-2020-322509] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 10/26/2020] [Accepted: 10/26/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Lipotoxic hepatocyte injury is a primary event in non-alcoholic steatohepatitis (NASH), but the mechanisms of lipotoxicity are not fully defined. Sphingolipids and free cholesterol (FC) mediate hepatocyte injury, but their link in NASH has not been explored. We examined the role of free cholesterol and sphingomyelin synthases (SMSs) that generate sphingomyelin (SM) and diacylglycerol (DAG) in hepatocyte pyroptosis, a specific form of programmed cell death associated with inflammasome activation, and NASH. DESIGN Wild-type C57BL/6J mice were fed a high fat and high cholesterol diet (HFHCD) to induce NASH. Hepatic SMS1 and SMS2 expressions were examined in various mouse models including HFHCD-fed mice and patients with NASH. Pyroptosis was estimated by the generation of the gasdermin-D N-terminal fragment. NASH susceptibility and pyroptosis were examined following knockdown of SMS1, protein kinase Cδ (PKCδ), or the NLR family CARD domain-containing protein 4 (NLRC4). RESULTS HFHCD increased the hepatic levels of SM and DAG while decreasing the level of phosphatidylcholine. Hepatic expression of Sms1 but not Sms2 was higher in mouse models and patients with NASH. FC in hepatocytes induced Sms1 expression, and Sms1 knockdown prevented HFHCD-induced NASH. DAG produced by SMS1 activated PKCδ and NLRC4 inflammasome to induce hepatocyte pyroptosis. Depletion of Nlrc4 prevented hepatocyte pyroptosis and the development of NASH. Conditioned media from pyroptotic hepatocytes activated the NOD-like receptor family pyrin domain containing 3 inflammasome (NLRP3) in Kupffer cells, but Nlrp3 knockout mice were not protected against HFHCD-induced hepatocyte pyroptosis. CONCLUSION SMS1 mediates hepatocyte pyroptosis through a novel DAG-PKCδ-NLRC4 axis and holds promise as a therapeutic target for NASH.
Collapse
Affiliation(s)
- Eun Hee Koh
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Ji Eun Yoon
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Myoung Seok Ko
- Biomedical Research Center, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jaechan Leem
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Ji-Young Yun
- Biomedical Research Center, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Chung Hwan Hong
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Yun Kyung Cho
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Seung Eun Lee
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jung Eun Jang
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Ji Yeon Baek
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Hyun Ju Yoo
- The Convergence Medicine Research Center, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Su Jung Kim
- The Convergence Medicine Research Center, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Chang Ohk Sung
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Joon Seo Lim
- Clinical Research Center, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Won-Il Jeong
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon, South Korea
| | - Sung Hoon Back
- School of Biological Sciences, University of Ulsan, Ulsan, South Korea
| | - In-Jeoung Baek
- The Convergence Medicine Research Center, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Sandra Torres
- Department of Cell Death and Proliferation, Instituto Investigaciones Biomédicas de Barcelona (IIBB), CSIC, Barcelona, Spain and Liver Unit-IDIBAPS and Centro de Investigación Biomédica en Red (CIBERehd), Barcelona, Spain
| | - Estel Solsona-Vilarrasa
- Department of Cell Death and Proliferation, Instituto Investigaciones Biomédicas de Barcelona (IIBB), CSIC, Barcelona, Spain and Liver Unit-IDIBAPS and Centro de Investigación Biomédica en Red (CIBERehd), Barcelona, Spain
| | - Laura Conde de la Rosa
- Department of Cell Death and Proliferation, Instituto Investigaciones Biomédicas de Barcelona (IIBB), CSIC, Barcelona, Spain and Liver Unit-IDIBAPS and Centro de Investigación Biomédica en Red (CIBERehd), Barcelona, Spain
| | - Carmen Garcia-Ruiz
- Department of Cell Death and Proliferation, Instituto Investigaciones Biomédicas de Barcelona (IIBB), CSIC, Barcelona, Spain and Liver Unit-IDIBAPS and Centro de Investigación Biomédica en Red (CIBERehd), Barcelona, Spain,University of Southern California Research Center for Alcoholic Liver and Pancreatic Diseases and Cirrhosis, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Ariel E Feldstein
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Jose C Fernandez-Checa
- Department of Cell Death and Proliferation, Instituto Investigaciones Biomédicas de Barcelona (IIBB), CSIC, Barcelona, Spain and Liver Unit-IDIBAPS and Centro de Investigación Biomédica en Red (CIBERehd), Barcelona, Spain .,University of Southern California Research Center for Alcoholic Liver and Pancreatic Diseases and Cirrhosis, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Ki-Up Lee
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| |
Collapse
|
135
|
Wu Y, Wang M, Yang T, Qin L, Hu Y, Zhao D, Wu L, Liu T. Cinnamic Acid Ameliorates Nonalcoholic Fatty Liver Disease by Suppressing Hepatic Lipogenesis and Promoting Fatty Acid Oxidation. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:9561613. [PMID: 34512784 PMCID: PMC8433026 DOI: 10.1155/2021/9561613] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/22/2021] [Accepted: 08/25/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Cinnamic acid (CA) has been shown to have many beneficial effects including regulating lipid metabolism and reducing obesity. However, its effect on nonalcoholic fatty liver disease (NAFDL) has not been investigated in detail. Thus, we performed this study in order to explore CA's effect on hepatic lipid metabolism and the underlying mechanisms. METHOD Oleic acid (OA) was used to induce lipid accumulation in HepG2 cells. After coincubation with CA, the cells were stained with oil red O and the triglyceride (TG) content was assessed. Key genes in lipogenesis and fatty acid oxidation pathways were tested. Additionally, db/db and wt/wt mice were divided into three groups, with the wt/wt mice representing the normal group and the db/db mice being divided into the NAFLD and CA groups. After 4 weeks of oral treatment, all mice were sacrificed and the blood lipid profile and liver tissues were assessed. RESULTS CA treatment reduced the lipid accumulation in HepG2 cells and in db/db mouse livers. ACLY, ACC, FAS, SCD1, PPARγ, and CD36 were significantly downregulated, while CPT1A, PGC1α, and PPARα were significantly upregulated. CONCLUSION CA's therapeutic effect on NAFLD may be attributed to its ability to lower hepatic lipid accumulation, which is mediated by suppression of hepatic lipogenesis and fatty acid intake, as well as increased fatty acid oxidation.
Collapse
Affiliation(s)
- You Wu
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing 100029, China
- Key Laboratory of Health Cultivation of Beijing, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Minghui Wang
- Chengdu Integrated TCM and Western Medicine Hospital, Chengdu 610016, China
| | - Tao Yang
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing 100029, China
- Key Laboratory of Health Cultivation of Beijing, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Lingling Qin
- Department of Science and Technology, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yaomu Hu
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing 100029, China
- Key Laboratory of Health Cultivation of Beijing, Beijing University of Chinese Medicine, Beijing 100029, China
- First School of Clinical Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Dan Zhao
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing 100029, China
- Key Laboratory of Health Cultivation of Beijing, Beijing University of Chinese Medicine, Beijing 100029, China
- First School of Clinical Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Lili Wu
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing 100029, China
- Key Laboratory of Health Cultivation of Beijing, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Tonghua Liu
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing 100029, China
- Key Laboratory of Health Cultivation of Beijing, Beijing University of Chinese Medicine, Beijing 100029, China
| |
Collapse
|
136
|
Sousa-Lima I, Kim HJ, Jones J, Kim YB. Rho-Kinase as a Therapeutic Target for Nonalcoholic Fatty Liver Diseases. Diabetes Metab J 2021; 45:655-674. [PMID: 34610720 PMCID: PMC8497927 DOI: 10.4093/dmj.2021.0197] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 08/25/2021] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a major public health problem and the most common form of chronic liver disease, affecting 25% of the global population. Although NAFLD is closely linked with obesity, insulin resistance, and type 2 diabetes mellitus, knowledge on its pathogenesis remains incomplete. Emerging data have underscored the importance of Rho-kinase (Rho-associated coiled-coil-containing kinase [ROCK]) action in the maintenance of normal hepatic lipid homeostasis. In particular, pharmacological blockade of ROCK in hepatocytes or hepatic stellate cells prevents the progression of liver diseases such as NAFLD and fibrosis. Moreover, mice lacking hepatic ROCK1 are protected against obesity-induced fatty liver diseases by suppressing hepatic de novo lipogenesis. Here we review the roles of ROCK as an indispensable regulator of obesity-induced fatty liver disease and highlight the key cellular pathway governing hepatic lipid accumulation, with focus on de novo lipogenesis and its impact on therapeutic potential. Consequently, a comprehensive understanding of the metabolic milieu linking to liver dysfunction triggered by ROCK activation may help identify new targets for treating fatty liver diseases such as NAFLD.
Collapse
Affiliation(s)
- Inês Sousa-Lima
- CEDOC-Chronic Disease Research Center, NOVA Medical School/ Faculty of Medical Sciences, New University of Lisbon, Lisbon, Portugal
| | - Hyun Jeong Kim
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - John Jones
- Center for Neuroscience and Cell Biology, University of Coimbra, Marquis of Pombal Square, Coimbra, Portugal
| | - Young-Bum Kim
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Corresponding author: Young-Bum Kim https://orcid.org/0000-0001-9471-6330 Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02215, USA E-mail:
| |
Collapse
|
137
|
Zhang Q, Lin W, Tian L, Di B, Yu J, Niu X, Liu J. Oxidized low-density lipoprotein activates extracellular signal-regulated kinase signaling to downregulate sortilin expression in liver sinusoidal endothelial cells. J Gastroenterol Hepatol 2021; 36:2610-2618. [PMID: 33694195 PMCID: PMC8518938 DOI: 10.1111/jgh.15486] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND AND AIM Both type 2 diabetes mellitus and non-alcoholic fatty liver disease are closely associated with elevated levels of low-density lipoprotein cholesterol and its oxidized form (ox-LDL). This study aimed to investigate the regulation of sortilin in liver tissue and its potential implications for lipid metabolism. METHODS Sixty male Wistar rats were randomly divided into four groups: control group (n = 15), ox-LDL group (n = 15), PD98059 group (n = 15), and ox-LDL + PD98059 group (n = 15). Liver sinusoidal endothelial cells were extracted from liver tissue of the control group and were identified using an anti-CD31 antibody. Lipid droplet accumulation was observed by Oil red O and hematoxylin-eosin staining. The protein expression levels were detected by immunohistochemical staining, real-time reverse transcription-polymerase chain reaction, and western blot. Histopathologic examinations were performed by Gomori methenamine silver staining. RESULTS The ox-LDL group exhibited increased lipid droplet accumulation. Further, ox-LDL activated the extracellular signal-regulated kinase (ERK)-mediated downregulation of sortilin expression, whereas blocking of ERK signaling by PD98059 increased sortilin protein expression. Consistently, hematoxylin-eosin staining showed that the structure of the hepatocytes was loose and disordered in arrangement, with lipid droplets present in the cytoplasm of the ox-LDL group. However, PD98059 significantly improved the integration of the scaffold structure. Gomori methenamine silver staining showed that the ox-LDL group had darker and more obvious fragmented silver nitrate deposits in the basement membrane and sinus space. CONCLUSIONS Sortilin can protect liver sinusoidal endothelial cells from injury and maintain integration of the liver scaffold structure in ox-LDL-induced lipid-injured liver.
Collapse
Affiliation(s)
- Qi Zhang
- Department of EndocrinologyGansu Provincial HospitalLanzhouChina,Clinical Research Center for Metabolic DiseasesGansu ProvincialLanzhouChina
| | - Wenyan Lin
- Gansu Provincial Hospital West CampusLanzhouChina,Clinical Research Center for Metabolic DiseasesGansu ProvincialLanzhouChina
| | - Limin Tian
- Department of EndocrinologyGansu Provincial HospitalLanzhouChina,Clinical Research Center for Metabolic DiseasesGansu ProvincialLanzhouChina
| | - Baoshan Di
- Gansu Provincial Hospital West CampusLanzhouChina
| | - Jing Yu
- Department of EndocrinologyGansu Provincial HospitalLanzhouChina,Clinical Research Center for Metabolic DiseasesGansu ProvincialLanzhouChina
| | - Xiang'e Niu
- Department of EndocrinologyGansu Provincial HospitalLanzhouChina,Clinical Research Center for Metabolic DiseasesGansu ProvincialLanzhouChina,Gansu University of Chinese MedicineLanzhouChina
| | - Jing Liu
- Department of EndocrinologyGansu Provincial HospitalLanzhouChina,Clinical Research Center for Metabolic DiseasesGansu ProvincialLanzhouChina
| |
Collapse
|
138
|
Pan MX, Zheng CY, Deng YJ, Tang KR, Nie H, Xie JQ, Liu DD, Tu GF, Yang QH, Zhang YP. Hepatic protective effects of Shenling Baizhu powder, a herbal compound, against inflammatory damage via TLR4/NLRP3 signalling pathway in rats with nonalcoholic fatty liver disease. JOURNAL OF INTEGRATIVE MEDICINE-JIM 2021; 19:428-438. [PMID: 34426178 DOI: 10.1016/j.joim.2021.07.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 05/18/2021] [Indexed: 02/06/2023]
Abstract
OBJECTIVE High-fat diet (HFD) and inflammation are two key contributors to nonalcoholic fatty liver disease (NAFLD). Shenling Baizhu powder (SLBZP), a classical herbal compound, has been successfully used to alleviate NAFLD. However, its specific mechanisms are not fully understood. In this study, we assessed the anti-NAFLD effect of SLBZP in vivo. METHODS Rats were fed an HFD with or without SLBZP or with probiotics. At the end of week 16, an echo magnetic resonance imaging (EchoMRI) body composition analyser was used to quantitatively analyse body composition; a micro-computed tomography (micro-CT) imaging system was used to evaluate whole body and liver fat; and the Moor full-field laser perfusion imager 2 was used to assess liver microcirculation, after which, all rats were sacrificed. Then, biochemical indicators in the blood and the ultrastructure of rat livers were evaluated. Protein expression related to the liver Toll-like receptor 4 (TLR4)/Nod-like receptor family pyrin domain-containing 3 (NLRP3) signalling pathway was assessed using Western blot analysis. Further, high-throughput screening of 29 related inflammatory factors in liver tissue was performed using a cytokine array. RESULTS SLBZP supplementation reduced body weight, serum free fatty acid, and insulin resistance index (P < 0.05). It also ameliorated liver microcirculation and ultrastructural abnormalities. EchoMRI and micro-CT quantitative analyses showed that treatment with SLBZP reduced fat mass and visceral fat (P < 0.05 and P < 0.01, respectively). In addition, SLBZP decreased the expression of lipopolysaccharide (LPS)-activated TLR4/NLRP3 signalling pathway-related proteins and altered the expression levels of some inflammatory cytokines in liver tissues. CONCLUSION SLBZP can inhibit NLRP3 inflammasome activation and interleukin-1β release by suppressing LPS-induced TLR4 expression in rats with HFD-induced NAFLD. Thus, SLBZP may be beneficial for the prevention and treatment of inflammatory damage and associated diseases.
Collapse
Affiliation(s)
- Mao-Xing Pan
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Chui-Yang Zheng
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Yuan-Jun Deng
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Kai-Rui Tang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Huan Nie
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Ji-Qian Xie
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Dong-Dong Liu
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Gui-Fang Tu
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Qin-He Yang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China.
| | - Yu-Pei Zhang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China.
| |
Collapse
|
139
|
Phytotherapy as Multi-Hit Therapy to Confront the Multiple Pathophysiology in Non-Alcoholic Fatty Liver Disease: A Systematic Review of Experimental Interventions. ACTA ACUST UNITED AC 2021; 57:medicina57080822. [PMID: 34441028 PMCID: PMC8400978 DOI: 10.3390/medicina57080822] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/02/2021] [Accepted: 08/10/2021] [Indexed: 02/06/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD), or metabolic dysfunction-associated fatty liver disease (MAFLD), is a metabolic condition distinguished by fat deposition in the hepatocytes. It has a prevalence of about 25% worldwide and is associated with other conditions such as diabetes mellitus, obesity, hypertension, etc. Background and Objectives: There is currently no approved drug therapy for NAFLD. Current measures in the management of NAFLD include lifestyle modification such as an increase in physical activity or weight loss. Development of NAFLD involves a number of parallel hits: including genetic predisposition, insulin resistance, disordered lipid metabolism, mitochondrial dysfunction, lipotoxicity, oxidative stress, etc. Herbal therapy may have a role to play in the treatment of NAFLD, due to their numerous bioactive constituents and the multiple pharmacological actions they exhibit. Therefore, this systematic review aims to investigate the potential multi-targeting effects of plant-derived extracts in experimental models of NAFLD. Materials and Methods: We performed a systematic search on databases and web search engines from the earliest available date to 30 April 2021, using relevant keywords. The study included articles published in English, assessing the effects of plant-derived extracts, fractions, or polyherbal mixtures in the treatment of NAFLD in animal models. These include their effects on at least disordered lipid metabolism, insulin resistance/type 2 diabetes mellitus (T2DM), and histologically confirmed steatosis with one or more of the following: oxidative stress, inflammation, hepatocyte injury, obesity, fibrosis, and cardiometabolic risks factors. Results: Nine articles fulfilled our inclusion criteria and the results demonstrated the ability of phytomedicines to simultaneously exert therapeutic actions on multiple targets related to NAFLD. Conclusions: These findings suggest that herbal extracts have the potential for effective treatment or management of NAFLD.
Collapse
|
140
|
Clifford BL, Sedgeman LR, Williams KJ, Morand P, Cheng A, Jarrett KE, Chan AP, Brearley-Sholto MC, Wahlström A, Ashby JW, Barshop W, Wohlschlegel J, Calkin AC, Liu Y, Thorell A, Meikle PJ, Drew BG, Mack JJ, Marschall HU, Tarling EJ, Edwards PA, de Aguiar Vallim TQ. FXR activation protects against NAFLD via bile-acid-dependent reductions in lipid absorption. Cell Metab 2021; 33:1671-1684.e4. [PMID: 34270928 PMCID: PMC8353952 DOI: 10.1016/j.cmet.2021.06.012] [Citation(s) in RCA: 170] [Impact Index Per Article: 56.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/12/2021] [Accepted: 06/21/2021] [Indexed: 12/24/2022]
Abstract
FXR agonists are used to treat non-alcoholic fatty liver disease (NAFLD), in part because they reduce hepatic lipids. Here, we show that FXR activation with the FXR agonist GSK2324 controls hepatic lipids via reduced absorption and selective decreases in fatty acid synthesis. Using comprehensive lipidomic analyses, we show that FXR activation in mice or humans specifically reduces hepatic levels of mono- and polyunsaturated fatty acids (MUFA and PUFA). Decreases in MUFA are due to FXR-dependent repression of Scd1, Dgat2, and Lpin1 expression, which is independent of SHP and SREBP1c. FXR-dependent decreases in PUFAs are mediated by decreases in lipid absorption. Replenishing bile acids in the diet prevented decreased lipid absorption in GSK2324-treated mice, suggesting that FXR reduces absorption via decreased bile acids. We used tissue-specific FXR KO mice to show that hepatic FXR controls lipogenic genes, whereas intestinal FXR controls lipid absorption. Together, our studies establish two distinct pathways by which FXR regulates hepatic lipids.
Collapse
Affiliation(s)
- Bethan L Clifford
- Department of Medicine, Division of Cardiology, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Leslie R Sedgeman
- Department of Medicine, Division of Cardiology, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Kevin J Williams
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA; Lipidomics Core Facility, Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Pauline Morand
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Angela Cheng
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Kelsey E Jarrett
- Department of Medicine, Division of Cardiology, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Alvin P Chan
- Department of Medicine, Division of Cardiology, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Madelaine C Brearley-Sholto
- Department of Medicine, Division of Cardiology, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Annika Wahlström
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Julianne W Ashby
- Department of Medicine, Division of Cardiology, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - William Barshop
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - James Wohlschlegel
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Anna C Calkin
- Lipid Metabolism & Cardiometabolic Disease Laboratory, Baker Heart & Diabetes Institute, Melbourne, VIC, Australia; Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Yingying Liu
- Lipid Metabolism & Cardiometabolic Disease Laboratory, Baker Heart & Diabetes Institute, Melbourne, VIC, Australia; Molecular Metabolism & Ageing Laboratory, Baker Heart & Diabetes Institute, Melbourne, VIC, Australia
| | - Anders Thorell
- Karolinska Institutet, Department of Clinical Science, Danderyd Hospital and Department of Surgery, Ersta Hospital, Stockholm, Sweden
| | - Peter J Meikle
- Metabolomics Laboratory, Baker Heart & Diabetes Institute, Melbourne, VIC, Australia
| | - Brian G Drew
- Central Clinical School, Monash University, Melbourne, VIC, Australia; Molecular Metabolism & Ageing Laboratory, Baker Heart & Diabetes Institute, Melbourne, VIC, Australia
| | - Julia J Mack
- Department of Medicine, Division of Cardiology, University of California, Los Angeles (UCLA), Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Hanns-Ulrich Marschall
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Elizabeth J Tarling
- Department of Medicine, Division of Cardiology, University of California, Los Angeles (UCLA), Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center (JCCC), UCLA, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Peter A Edwards
- Department of Medicine, Division of Cardiology, University of California, Los Angeles (UCLA), Los Angeles, CA, USA; Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Thomas Q de Aguiar Vallim
- Department of Medicine, Division of Cardiology, University of California, Los Angeles (UCLA), Los Angeles, CA, USA; Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center (JCCC), UCLA, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles (UCLA), Los Angeles, CA, USA.
| |
Collapse
|
141
|
Ding Y, Li G, Zhou Z, Deng T. Molecular mechanisms underlying hepatitis C virus infection-related diabetes. Metabolism 2021; 121:154802. [PMID: 34090869 DOI: 10.1016/j.metabol.2021.154802] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/27/2021] [Accepted: 05/31/2021] [Indexed: 12/16/2022]
Abstract
Diabetes is a noncommunicable widespread disease that poses the risk of severe complications in patients, with certain complications being life-threatening. Hepatitis C is an infectious disease that mainly causes liver damage, which is also a profound threat to human health. Hepatitis C virus (HCV) infection has many extrahepatic manifestations, including diabetes. Multiple mechanisms facilitate the strong association between HCV and diabetes. HCV infection can affect the insulin signaling pathway in liver and pancreatic tissue and change the profiles of circulating microRNAs, which may further influence the occurrence and development of diabetes. This review describes how HCV infection causes diabetes and discusses the current research progress with respect to HCV infection-related diabetes.
Collapse
Affiliation(s)
- Yujin Ding
- National Clinical Research Center for Metabolic Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China; Key Laboratory of Diabetes Immunology, Ministry of Education, and Metabolic Syndrome Research Center, The Second Xiangya Hospital of Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Guangdi Li
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha 410011, Hunan, China
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China; Key Laboratory of Diabetes Immunology, Ministry of Education, and Metabolic Syndrome Research Center, The Second Xiangya Hospital of Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Tuo Deng
- National Clinical Research Center for Metabolic Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China; Key Laboratory of Diabetes Immunology, Ministry of Education, and Metabolic Syndrome Research Center, The Second Xiangya Hospital of Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China; Clinical Immunology Center, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China.
| |
Collapse
|
142
|
Behl T, Gupta A, Sehgal A, Sharma S, Singh S, Sharma N, Diaconu CC, Rahdar A, Hafeez A, Bhatia S, Al-Harrasi A, Bungau S. A spotlight on underlying the mechanism of AMPK in diabetes complications. Inflamm Res 2021; 70:939-957. [PMID: 34319417 DOI: 10.1007/s00011-021-01488-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/29/2021] [Accepted: 06/08/2021] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE Type 2 diabetes (T2D) is one of the centenarian metabolic disorders and is considered as a stellar and leading health issue worldwide. According to the International Diabetes Federation (IDF) Diabetes Atlas and National Diabetes Statistics, the number of diabetic patients will increase at an exponential rate from 463 to 700 million by the year 2045. Thus, there is a great need for therapies targeting functions that can help in maintaining the homeostasis of glucose levels and improving insulin sensitivity. 5' adenosine monophosphate-activated protein kinase (AMPK) activation, by various direct and indirect factors, might help to overcome the hurdles (like insulin resistance) associated with the conventional approach. MATERIALS AND RESULTS A thorough review and analysis was conducted using various database including MEDLINE and EMBASE databases, with Google scholar using various keywords. This extensive review concluded that various drugs (plant-based, synthetic indirect/direct activators) are available, showing tremendous potential in maintaining the homeostasis of glucose and lipid metabolism, without causing insulin resistance, and improving insulin sensitivity. Moreover, these drugs have an effect against diabetes and are therapeutically beneficial in the treatment of diabetes-associated complications (neuropathy and nephropathy) via mechanism involving inhibition of nuclear translocation of SMAD4 (SMAD family member) expression and association with peripheral nociceptive neurons mediated by AMPK. CONCLUSION From the available information, it may be concluded that various indirect/direct activators show tremendous potential in maintaining the homeostasis of glucose and lipid metabolism, without resulting in insulin resistance, and may improve insulin sensitivity, as well. Therefore, in a nut shell, it may be concluded that the regulation of APMK functions by various direct/indirect activators may bring promising results. These activators may emerge as a novel therapy in diabetes and its associated complications.
Collapse
Affiliation(s)
- Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Amit Gupta
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Sanchay Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Camelia Cristina Diaconu
- Internal Medicine Department, Clinical Emergency Hospital of Bucharest, Bucharest, Romania.,Department 5, 'Carol Davila' University of Medicine and Pharmacy, Bucharest, Romania
| | - Abbas Rahdar
- Department of Physics, University of Zabol, Zabol, Iran
| | - Abdul Hafeez
- Glocal School of Pharmacy, Glocal University, Mirzapur, Uttar Pradesh, India
| | - Saurabh Bhatia
- Amity Institute of Pharmacy, Amity University, Haryana, India.,Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Oman
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Oman
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
143
|
von Loeffelholz C, Coldewey SM, Birkenfeld AL. A Narrative Review on the Role of AMPK on De Novo Lipogenesis in Non-Alcoholic Fatty Liver Disease: Evidence from Human Studies. Cells 2021; 10:cells10071822. [PMID: 34359991 PMCID: PMC8306246 DOI: 10.3390/cells10071822] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/01/2021] [Accepted: 07/15/2021] [Indexed: 02/06/2023] Open
Abstract
5′AMP-activated protein kinase (AMPK) is known as metabolic sensor in mammalian cells that becomes activated by an increasing adenosine monophosphate (AMP)/adenosine triphosphate (ATP) ratio. The heterotrimeric AMPK protein comprises three subunits, each of which has multiple phosphorylation sites, playing an important role in the regulation of essential molecular pathways. By phosphorylation of downstream proteins and modulation of gene transcription AMPK functions as a master switch of energy homeostasis in tissues with high metabolic turnover, such as the liver, skeletal muscle, and adipose tissue. Regulation of AMPK under conditions of chronic caloric oversupply emerged as substantial research target to get deeper insight into the pathogenesis of non-alcoholic fatty liver disease (NAFLD). Evidence supporting the role of AMPK in NAFLD is mainly derived from preclinical cell culture and animal studies. Dysbalanced de novo lipogenesis has been identified as one of the key processes in NAFLD pathogenesis. Thus, the scope of this review is to provide an integrative overview of evidence, in particular from clinical studies and human samples, on the role of AMPK in the regulation of primarily de novo lipogenesis in human NAFLD.
Collapse
Affiliation(s)
- Christian von Loeffelholz
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, 07747 Jena, Germany;
- Correspondence: ; Tel.: +49-3641-9323-177; Fax: +49-3641-9323-102
| | - Sina M. Coldewey
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, 07747 Jena, Germany;
- Septomics Research Center, Jena University Hospital, 07747 Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, 07747 Jena, Germany
| | - Andreas L. Birkenfeld
- Department of Diabetology Endocrinology and Nephrology, University Hospital Tübingen, Eberhard Karls University Tübingen, 72074 Tübingen, Germany;
- Department of Therapy of Diabetes, Institute of Diabetes Research and Metabolic Diseases in the Helmholtz Center Munich, Eberhard Karls University Tübingen, 72074 Tübingen, Germany
- Division of Diabetes and Nutritional Sciences, Rayne Institute, King’s College London, London SE5 9RJ, UK
| |
Collapse
|
144
|
Kirilmaz OB, Salegaonkar AR, Shiau J, Uzun G, Ko HS, Lee HF, Park S, Kwon G. Study of blood glucose and insulin infusion rate in real-time in diabetic rats using an artificial pancreas system. PLoS One 2021; 16:e0254718. [PMID: 34270619 PMCID: PMC8284668 DOI: 10.1371/journal.pone.0254718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 07/01/2021] [Indexed: 11/19/2022] Open
Abstract
Artificial pancreas system (APS) is an emerging new treatment for type 1 diabetes mellitus. The aim of this study was to develop a rat APS as a research tool and demonstrate its application. We established a rat APS using Medtronic Minimed Pump 722, Medtronic Enlite sensor, and the open artificial pancreas system as a controller. We tested different dilutions of Humalog (100 units/ml) in saline ranged from 1:3 to 1:20 and determined that 1:7 dilution works well for rats with ~500g bodyweight. Blood glucose levels (BGL) of diabetic rats fed with chow diet (58% carbohydrate) whose BGL was managed by the closed-loop APS for the total duration of 207h were in euglycemic range (70-180 mg/dl) for 94.5% of the time with 2.1% and 3.4% for hyperglycemia (>180mg/dl) and hypoglycemia (<70 mg/dl), respectively. Diabetic rats fed with Sucrose pellets (94.8% carbohydrate) for the experimental duration of 175h were in euglycemic range for 61% of the time with 35% and 4% for hyperglycemia and hypoglycemia, respectively. Heathy rats fed with chow diet showed almost a straight line of BGL ~ 95 mg/dl (average 94.8 mg/dl) during the entire experimental period (281h), which was minimally altered by food intake. In the healthy rats, feeding sucrose pellets caused greater range of BGL in high and low levels but still within euglycemic range (99.9%). Next, to study how healthy and diabetic rats handle supra-physiological concentrations of glucose, we intraperitoneally injected various amounts of 50% dextrose (2, 3, 4g/kg) and monitored BGL. Duration of hyperglycemia after injection of 50% dextrose at all three different concentrations was significantly greater for healthy rats than diabetic rats, suggesting that insulin infusion by APS was superior in reducing BGL as compared to natural insulin released from pancreatic β-cells. Ex vivo studies showed that islets isolated from diabetic rats were almost completely devoid of pancreatic β-cells but with intact α-cells as expected. Lipid droplet deposition in the liver of diabetic rats was significantly lower with higher levels of triacylglyceride in the blood as compared to those of healthy rats, suggesting lipid metabolism was altered in diabetic rats. However, glycogen storage in the liver determined by Periodic acid-Schiff staining was not altered in diabetic rats as compared to healthy rats. A rat APS may be used as a powerful tool not only to study alterations of glucose and insulin homeostasis in real-time caused by diet, exercise, hormones, or antidiabetic agents, but also to test mathematical and engineering models of blood glucose prediction or new algorithms for closed-loop APS.
Collapse
MESH Headings
- Animals
- Blood Glucose/analysis
- Blood Glucose/drug effects
- Diabetes Mellitus, Experimental/blood
- Diabetes Mellitus, Experimental/chemically induced
- Diabetes Mellitus, Experimental/diagnosis
- Diabetes Mellitus, Experimental/therapy
- Diabetes Mellitus, Type 1/blood
- Diabetes Mellitus, Type 1/chemically induced
- Diabetes Mellitus, Type 1/diagnosis
- Diabetes Mellitus, Type 1/therapy
- Glycated Hemoglobin/analysis
- Humans
- Infusions, Intravenous/instrumentation
- Infusions, Intravenous/methods
- Insulin/administration & dosage
- Male
- Pancreas, Artificial
- Rats
- Streptozocin/administration & dosage
- Streptozocin/toxicity
Collapse
Affiliation(s)
- Omer Batuhan Kirilmaz
- School of Engineering, Southern Illinois University Edwardsville, Edwardsville, Illinois, United States of America
| | | | - Justin Shiau
- School of Pharmacy, Southern Illinois University Edwardsville, Edwardsville, Illinois, United States of America
| | - Guney Uzun
- School of Engineering, Southern Illinois University Edwardsville, Edwardsville, Illinois, United States of America
| | - Hoo Sang Ko
- School of Engineering, Southern Illinois University Edwardsville, Edwardsville, Illinois, United States of America
| | - H. Felix Lee
- School of Engineering, Southern Illinois University Edwardsville, Edwardsville, Illinois, United States of America
| | - Sarah Park
- Research and Instructional Services, Duke University, Durham, North Carolina, United States of America
| | - Guim Kwon
- School of Pharmacy, Southern Illinois University Edwardsville, Edwardsville, Illinois, United States of America
- * E-mail:
| |
Collapse
|
145
|
Schumann T, König J, von Loeffelholz C, Vatner DF, Zhang D, Perry RJ, Bernier M, Chami J, Henke C, Kurzbach A, El-Agroudy NN, Willmes DM, Pesta D, de Cabo R, O Sullivan JF, Simon E, Shulman GI, Hamilton BS, Birkenfeld AL. Deletion of the diabetes candidate gene Slc16a13 in mice attenuates diet-induced ectopic lipid accumulation and insulin resistance. Commun Biol 2021; 4:826. [PMID: 34211098 PMCID: PMC8249653 DOI: 10.1038/s42003-021-02279-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 05/20/2021] [Indexed: 12/12/2022] Open
Abstract
Genome-wide association studies have identified SLC16A13 as a novel susceptibility gene for type 2 diabetes. The SLC16A13 gene encodes SLC16A13/MCT13, a member of the solute carrier 16 family of monocarboxylate transporters. Despite its potential importance to diabetes development, the physiological function of SLC16A13 is unknown. Here, we validate Slc16a13 as a lactate transporter expressed at the plasma membrane and report on the effect of Slc16a13 deletion in a mouse model. We show that Slc16a13 increases mitochondrial respiration in the liver, leading to reduced hepatic lipid accumulation and increased hepatic insulin sensitivity in high-fat diet fed Slc16a13 knockout mice. We propose a mechanism for improved hepatic insulin sensitivity in the context of Slc16a13 deficiency in which reduced intrahepatocellular lactate availability drives increased AMPK activation and increased mitochondrial respiration, while reducing hepatic lipid content. Slc16a13 deficiency thereby attenuates hepatic diacylglycerol-PKCε mediated insulin resistance in obese mice. Together, these data suggest that SLC16A13 is a potential target for the treatment of type 2 diabetes and non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Tina Schumann
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Jörg König
- Clinical Pharmacology and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | | | - Daniel F Vatner
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Dongyan Zhang
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Rachel J Perry
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Michel Bernier
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Jason Chami
- Heart Research Institute, Newtown, NSW, Australia
| | - Christine Henke
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Anica Kurzbach
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Nermeen N El-Agroudy
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Diana M Willmes
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Dominik Pesta
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
- Centre for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Rafael de Cabo
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - John F O Sullivan
- Heart Research Institute, Newtown, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Eric Simon
- Computational Biology, Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Gerald I Shulman
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Bradford S Hamilton
- CardioMetabolic Diseases Research, Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Andreas L Birkenfeld
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine, Technische Universität Dresden, Dresden, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- King's College London, Department of Diabetes, School of Life Course Science, London, UK.
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany.
- Department of Endocrinology, Diabetology and Nephrology, University Hospital of Tübingen, Tübingen, Germany.
| |
Collapse
|
146
|
Luo H, Zhao M, Feng X, Gao X, Hong M, Liu M, Li Y, Liu W, Liu Y, Yu C, Cao Y, Yang X, Fang Z, Zhang P. Decreased plasma n6 : n3 polyunsaturated fatty acids ratio interacting with high C-peptide promotes non-alcoholic fatty liver disease in type 2 diabetes patients. J Diabetes Investig 2021; 12:1263-1271. [PMID: 33244871 PMCID: PMC8264392 DOI: 10.1111/jdi.13469] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/28/2020] [Accepted: 11/12/2020] [Indexed: 12/13/2022] Open
Abstract
AIMS/INTRODUCTION To explore relationships between polyunsaturated fatty acids (PUFA) and non-alcoholic fatty liver disease (NAFLD) in patients with type 2 diabetes, and whether insulin action has an interactive effect with PUFA on NAFLD progression. MATERIALS AND METHODS We extracted clinical and omics data of 482 type 2 diabetes patients from a tertiary hospital consecutively from April 2018 to April 2019. NAFLD was estimated by ultrasound at admission. Plasma fasting n3 and n6 fatty acids were quantified by liquid chromatography-tandem mass spectrometry analysis. Restricted cubic spline nested in binary logistic regression was used to select the cut-off point, and estimate odds ratios and 95% confidence intervals. Additive interactions of the n6 : n3 ratio with insulin action for NAFLD were estimated using relative excess risk due to interaction, attributable proportion due to interaction and synergy index. Relative excess risk due to interaction >0, attributable proportion due to interaction >0 or synergy index >1 indicates biological interaction. Spearman correlation analysis was used to obtain partial correlation coefficients between PUFA and hallmarks of NAFLD. RESULTS Of 482 patients, 313 were with and 169 were without NAFLD. N3 ≥800 and n6 PUFA ≥8,100 μmol/L were independently associated with increased NAFLD risk; n6 : n3 ratio ≤10 was associated with NAFLD (odds ratio 1.80, 95% confidence interval 1.20-2.71), and the effect size was amplified by high C-peptide (odds ratio 8.89, 95% confidence interval 4.48-17.7) with significant interaction. The additive interaction of the n6 : n3 ratio and fasting insulin was not significant. CONCLUSION Decreased n6 : n3 ratio was associated with increased NAFLD risk in type 2 diabetes patients, and the effect was only significant and amplified when there was the co-presence of high C-peptide.
Collapse
Affiliation(s)
- Hui‐Huan Luo
- Department of EndocrinologyThe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
- Department of Toxicology and Sanitary ChemistrySchool of Public HealthTianjin Medical UniversityTianjinChina
| | - Meng‐Di Zhao
- Department of EndocrinologyThe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Xiao‐Fei Feng
- Department of Toxicology and Sanitary ChemistrySchool of Public HealthTianjin Medical UniversityTianjinChina
| | - Xiao‐Qian Gao
- Department of Toxicology and Sanitary ChemistrySchool of Public HealthTianjin Medical UniversityTianjinChina
| | - Mo Hong
- RSKT Biopharma IncDalianLiaoningChina
| | | | - Yan‐Ping Li
- Department of EndocrinologyThe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Wan‐Qiu Liu
- Department of EndocrinologyThe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Yu‐Mo Liu
- Department of EndocrinologyThe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Cheng‐Cheng Yu
- Department of EndocrinologyThe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Yun‐Feng Cao
- RSKT Biopharma IncDalianLiaoningChina
- Dalian Institute of Chemical PhysicsChinese Academy of SciencesBeijingChina
| | - Xi‐Lin Yang
- Department of Epidemiology and BiostatisticsSchool of Public HealthTianjin Medical UniversityTianjinChina
| | - Zhong‐Ze Fang
- Department of Toxicology and Sanitary ChemistrySchool of Public HealthTianjin Medical UniversityTianjinChina
| | - Ping Zhang
- Department of EndocrinologyThe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
| |
Collapse
|
147
|
Lim S, Kim JW, Targher G. Links between metabolic syndrome and metabolic dysfunction-associated fatty liver disease. Trends Endocrinol Metab 2021; 32:500-514. [PMID: 33975804 DOI: 10.1016/j.tem.2021.04.008] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/15/2021] [Accepted: 04/15/2021] [Indexed: 02/08/2023]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) is a chronic condition characterized by hepatic fat accumulation combined with underlying metabolic dysregulation. Having evolved from the previous term of nonalcoholic fatty liver disease (NAFLD), the term MAFLD more closely implicates the presence of overweight/obesity, type 2 diabetes, or metabolic dysregulation as essential pathogenic factors, leading to better identification of individuals with this metabolic liver disease. Low-grade inflammation, increased oxidative stress, mitochondrial dysfunction, and intestinal dysbiosis are also involved in its pathogenesis. MAFLD is not only associated with liver-related complications, but also with adverse cardiometabolic outcomes. Further studies are needed to assess whether the newly proposed definition of MAFLD is more accurate than the NAFLD in predicting the adverse liver-related and extrahepatic outcomes.
Collapse
Affiliation(s)
- Soo Lim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seoul, Korea.
| | - Jin-Wook Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seoul, Korea
| | - Giovanni Targher
- Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Verona, Verona, Italy.
| |
Collapse
|
148
|
Qian H, Chao X, Williams J, Fulte S, Li T, Yang L, Ding WX. Autophagy in liver diseases: A review. Mol Aspects Med 2021; 82:100973. [PMID: 34120768 DOI: 10.1016/j.mam.2021.100973] [Citation(s) in RCA: 155] [Impact Index Per Article: 51.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 05/29/2021] [Accepted: 05/30/2021] [Indexed: 02/07/2023]
Abstract
The liver is a highly dynamic metabolic organ that plays critical roles in plasma protein synthesis, gluconeogenesis and glycogen storage, cholesterol metabolism and bile acid synthesis as well as drug/xenobiotic metabolism and detoxification. Research from the past decades indicate that autophagy, the cellular catabolic process mediated by lysosomes, plays an important role in maintaining cellular and metabolic homeostasis in the liver. Hepatic autophagy fluctuates with hormonal cues and the availability of nutrients that respond to fed and fasting states as well as circadian activities. Dysfunction of autophagy in liver parenchymal and non-parenchymal cells can lead to various liver diseases including non-alcoholic fatty liver diseases, alcohol associated liver disease, drug-induced liver injury, cholestasis, viral hepatitis and hepatocellular carcinoma. Therefore, targeting autophagy may be a potential strategy for treating these various liver diseases. In this review, we will discuss the current progress on the understanding of autophagy in liver physiology. We will also discuss several forms of selective autophagy in the liver and the molecular signaling pathways in regulating autophagy of different cell types and their implications in various liver diseases.
Collapse
Affiliation(s)
- Hui Qian
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS, 66160, USA
| | - Xiaojuan Chao
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS, 66160, USA
| | - Jessica Williams
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS, 66160, USA
| | - Sam Fulte
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS, 66160, USA
| | - Tiangang Li
- Harold Hamm Diabetes Center, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Ling Yang
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS, 66160, USA.
| |
Collapse
|
149
|
Raza S, Tewari A, Rajak S, Sinha RA. Vitamins and non-alcoholic fatty liver disease: A Molecular Insight ⋆. LIVER RESEARCH 2021; 5:62-71. [PMID: 34221537 PMCID: PMC7611112 DOI: 10.1016/j.livres.2021.03.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The incidence of non-alcoholic fatty liver disease (NAFLD) is rising rapidly across the globe. NAFLD pathogenesis is largely driven by an imbalance in hepatic energy metabolism and at present, there is no approved drug for its treatment. The liver plays a crucial role in micronutrient metabolism and deregulation of this micronutrient metabolism may contribute to the pathogenesis of NAFLD. Vitamins regulate several enzymatic processes in the liver, and derangement in vitamin metabolism is believed to play a critical role in NAFLD progression. The anti-oxidant activities of vitamin C and E have been attributed to mitigate hepatocyte injury, and alterations in the serum levels of vitamin D, vitamin B12 and folate have shown a strong correlation with NAFLD severity. This review aims to highlight the role of these vitamins, which represent promising therapeutic targets for the management of NAFLD.
Collapse
Affiliation(s)
- Sana Raza
- Corresponding author: Dr. Rohit A. Sinha (), Dr. Sana Raza ()
| | | | | | - Rohit A. Sinha
- Corresponding author: Dr. Rohit A. Sinha (), Dr. Sana Raza ()
| |
Collapse
|
150
|
Won G, Choi SI, Kang CH, Kim GH. Lactiplantibacillus plantarum MG4296 and Lacticaseibacillus paracasei MG5012 Ameliorates Insulin Resistance in Palmitic Acid-Induced HepG2 Cells and High Fat Diet-Induced Mice. Microorganisms 2021; 9:1139. [PMID: 34070604 PMCID: PMC8228052 DOI: 10.3390/microorganisms9061139] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/21/2021] [Accepted: 05/21/2021] [Indexed: 12/30/2022] Open
Abstract
The purpose of this study was to evaluate the capacity of Lactiplantibacillus plantarum MG4296 (MG4296) and Lacticaseibacillus paracasei MG5012 (MG5012) on insulin resistance (IR) and diabetes-related metabolic changes in palmitic acid (PA)-induced HepG2 cells and high-fat diet-induced mice. In vitro, cell-free extracts of MG4296 and MG5012 alleviated IR by increasing glucose uptake and glycogen content in PA-induced insulin-resistant HepG2 cells. In vivo, MG4296 and MG5012 supplementation markedly decreased body weight and glucose tolerance. Administration of both strains also improved serum glucose, glycated hemoglobin, insulin, triglyceride, LDL/HDL ratio, and homeostatic model assessment of IR (HOMA-IR). Histopathological analysis of liver tissue demonstrated a significant reduction in lipid accumulation and glycogen content. Moreover, MG4296 and MG5012 treatment enhanced phosphoinositide-3 kinase (PI3K)/protein kinase B (Akt) expression in the liver. Overall, MG4296 and MG5012 could prevent HFD-induced glucose tolerance and hyperglycemia by improving IR. Therefore, L. plantarum MG4296 and L. paracasei MG5012 could be useful as new probiotics candidates to improve T2DM.
Collapse
Affiliation(s)
- Gayeong Won
- Department of Health Functional New Materials, Duksung Women’s University, Seoul 01369, Korea;
| | - Soo-Im Choi
- Department of Health Functional New Materials, Duksung Women’s University, Seoul 01369, Korea;
| | | | - Gun-Hee Kim
- Department of Food and Nutrition, Duksung Women’s University, Seoul 01369, Korea
| |
Collapse
|