51
|
Aroner SA, Yang M, Li J, Furtado JD, Sacks FM, Tjønneland A, Overvad K, Cai T, Jensen MK. Apolipoprotein C-III and High-Density Lipoprotein Subspecies Defined by Apolipoprotein C-III in Relation to Diabetes Risk. Am J Epidemiol 2017; 186:736-744. [PMID: 28520887 DOI: 10.1093/aje/kwx143] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 11/09/2016] [Indexed: 12/19/2022] Open
Abstract
Apolipoprotein C-III (apoC-III) is a potentially novel biomarker that may play an important role in the pathogenesis of diabetes, particularly when present on the surface of high-density lipoprotein (HDL). In a case-cohort study carried out among 434 incident diabetes cases occurring before 2007 and 3,101 noncases in the Danish Diet, Cancer, and Health Study, we examined associations of baseline (1993-1997) plasma concentrations of apoC-III and subspecies of HDL defined by the presence or absence of apoC-III with risk of diabetes using Cox regression. ApoC-III was strongly associated with risk of diabetes (for top quintile vs. bottom quintile, hazard ratio (HR) = 3.43, 95% confidence interval (CI): 1.75, 6.70; P-trend < 0.001). The cholesterol concentration of HDL (HDL cholesterol (HDL-C)) without apoC-III was inversely associated with risk of diabetes (HR = 0.48, 95% CI: 0.27, 0.85; P-trend = 0.002), more so than total HDL-C (HR = 0.60, 95% CI: 0.35, 1.03; P-trend = 0.04), whereas HDL-C with apoC-III was not associated (HR = 1.05, 95% CI: 0.50, 2.21; P-trend = 0.44) (for HDL-C with apoC-III vs. HDL-C without apoC-III, P-heterogeneity = 0.002). ApoC-III itself is a strong risk marker for diabetes, and its presence on HDL may impair the antidiabetogenic properties of HDL. ApoC-III has potential to be a therapeutic target for the prevention of diabetes.
Collapse
|
52
|
Insulin Resistance, Obesity and Lipotoxicity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 960:277-304. [PMID: 28585204 DOI: 10.1007/978-3-319-48382-5_12] [Citation(s) in RCA: 285] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Lipotoxicity , originally used to describe the destructive effects of excess fat accumulation on glucose metabolism, causes functional impairments in several metabolic pathways, both in adipose tissue and peripheral organs, like liver, heart, pancreas and muscle. Lipotoxicity has roles in insulin resistance and pancreatic beta cell dysfunction. Increased circulating levels of lipids and the metabolic alterations in fatty acid utilization and intracellular signaling, have been related to insulin resistance in muscle and liver. Different pathways, like novel protein kinase c pathways and the JNK-1 pathway are involved as the mechanisms of how lipotoxicity leads to insulin resistance in nonadipose tissue organs, such as liver and muscle. Mitochondrial dysfunction plays a role in the pathogenesis of insulin resistance. Endoplasmic reticulum stress, through mainly increased oxidative stress, also plays important role in the etiology of insulin resistance, especially seen in non-alcoholic fatty liver disease. Visceral adiposity and insulin resistance both increase the cardiometabolic risk and lipotoxicity seems to play a crucial role in the pathophysiology of these associations.
Collapse
|
53
|
Sundaram M, Curtis KR, Amir Alipour M, LeBlond ND, Margison KD, Yaworski RA, Parks RJ, McIntyre AD, Hegele RA, Fullerton MD, Yao Z. The apolipoprotein C-III (Gln38Lys) variant associated with human hypertriglyceridemia is a gain-of-function mutation. J Lipid Res 2017; 58:2188-2196. [PMID: 28887372 DOI: 10.1194/jlr.m077313] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 09/04/2017] [Indexed: 11/20/2022] Open
Abstract
Recent cell culture and animal studies have suggested that expression of human apo C-III in the liver has a profound impact on the triacylglycerol (TAG)-rich VLDL1 production under lipid-rich conditions. The apoC-III Gln38Lys variant was identified in subjects of Mexican origin with moderate hypertriglyceridemia. We postulated that Gln38Lys (C3QK), being a gain-of-function mutation, promotes hepatic VLDL1 assembly/secretion. To test this hypothesis, we expressed C3QK in McA-RH7777 cells and apoc3-null mice to contrast its effect with WT apoC-III (C3WT). In both model systems, C3QK expression increased the secretion of VLDL1-TAG (by 230%) under lipid-rich conditions. Metabolic labeling experiments with C3QK cells showed an increase in de novo lipogenesis (DNL). Fasting plasma concentration of TAG, cholesterol, cholesteryl ester, and FA were increased in C3QK mice as compared with C3WT mice. Liver of C3QK mice also displayed an increase in DNL and expression of lipogenic genes as compared with that in C3WT mice. These results suggest that C3QK variant is a gain-of-function mutation that can stimulate VLDL1 production, through enhanced DNL.
Collapse
Affiliation(s)
- Meenakshi Sundaram
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Kaitlin R Curtis
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Mohsen Amir Alipour
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Nicholas D LeBlond
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Kaitlyn D Margison
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Rebecca A Yaworski
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Robin J Parks
- Ottawa Hospital Research Institute Ottawa, Ontario K1H 8L6, Canada
| | - Adam D McIntyre
- Department of Medicine and Robarts Research Institute, Schulich School of Medicine, Western University, London, Ontario N6A 5B7, Canada
| | - Robert A Hegele
- Department of Medicine and Robarts Research Institute, Schulich School of Medicine, Western University, London, Ontario N6A 5B7, Canada
| | - Morgan D Fullerton
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Zemin Yao
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| |
Collapse
|
54
|
A Novel Role for RARα Agonists as Apolipoprotein CIII Inhibitors Identified from High Throughput Screening. Sci Rep 2017; 7:5824. [PMID: 28724938 PMCID: PMC5517646 DOI: 10.1038/s41598-017-05163-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 05/25/2017] [Indexed: 01/23/2023] Open
Abstract
Elevated triglyceride (TG) levels are well-correlated with the risk for cardiovascular disease (CVD). Apolipoprotein CIII (ApoC-III) is a key regulator of plasma TG levels through regulation of lipolysis and lipid synthesis. To identify novel regulators of TG levels, we carried out a high throughput screen (HTS) using an ApoC-III homogenous time resolved fluorescence (HTRF) assay. We identified several retinoic acid receptor (RAR) agonists that reduced secreted ApoC-III levels in human hepatic cell lines. The RARα specific agonist AM580 inhibited secreted ApoC-III by >80% in Hep3B cells with an EC50 ~2.9 nM. In high-fat diet induced fatty-liver mice, AM580 reduced ApoC-III levels in liver as well as in plasma (~60%). In addition, AM580 treatment effectively reduced body weight, hepatic and plasma TG, and total cholesterol (TC) levels. Mechanistically, AM580 suppresses ApoC-III synthesis by downregulation of HNF4α and upregulation of SHP1 expression. Collectively, these studies suggest that an RARα specific agonist may afford a new strategy for lipid-lowering and CVD risk reduction.
Collapse
|
55
|
Petersen MC, Shulman GI. Roles of Diacylglycerols and Ceramides in Hepatic Insulin Resistance. Trends Pharmacol Sci 2017; 38:649-665. [PMID: 28551355 DOI: 10.1016/j.tips.2017.04.004] [Citation(s) in RCA: 245] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 04/19/2017] [Accepted: 04/20/2017] [Indexed: 12/22/2022]
Abstract
Although ample evidence links hepatic lipid accumulation with hepatic insulin resistance, the mechanistic basis of this association is incompletely understood and controversial. Diacylglycerols (DAGs) and ceramides have emerged as the two best-studied putative mediators of lipid-induced hepatic insulin resistance. Both lipids were first associated with insulin resistance in skeletal muscle and were subsequently hypothesized to mediate insulin resistance in the liver. However, the putative roles for DAGs and ceramides in hepatic insulin resistance have proved more complex than originally imagined, with various genetic and pharmacologic manipulations yielding a vast and occasionally contradictory trove of data to sort. In this review we examine the state of this field, turning a critical eye toward both DAGs and ceramides as putative mediators of lipid-induced hepatic insulin resistance.
Collapse
Affiliation(s)
- Max C Petersen
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Gerald I Shulman
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA; Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
56
|
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common and important chronic liver disease in the world. As the prevalence of obesity increases in adults and children, the incidence of NAFLD has increased rapidly, reaching 17% to 33%. NAFLD is clinically divided into two forms: simple fatty liver (SFL) and non-alcoholic steatohepatitis (NASH), with NASH accounting for 1/3-1/2 of all NAFLD cases. The probability of developing cirrhosis is 0.6%-3.0% in patients with SFL for 10-20 years, and as high as 15%-25% in patients with NASH for 10-15 years. Approximately 1% of cirrhosis cases develop hepatocellular carcinoma each year. The pathogenesis of NAFLD is still not completely clear. It is generally believed that age, sex, obesity, insulin resistance, cytokines, gene polymorphism, and intestinal microflora are involved in the pathogenesis of NAFLD. An in-depth understanding of the pathogenesis of NAFLD can provide a basis for treatment of this disease. In recent years, cytokines or genes have been reported as targets for NAFLD treatment with appreciated effects. Since there is currently no specific treatment for NAFLD, targeted therapy may have a profound impact on the prognosis of the disease.
Collapse
|
57
|
Cheng X, Yamauchi J, Lee S, Zhang T, Gong Z, Muzumdar R, Qu S, Dong HH. APOC3 Protein Is Not a Predisposing Factor for Fat-induced Nonalcoholic Fatty Liver Disease in Mice. J Biol Chem 2017; 292:3692-3705. [PMID: 28115523 DOI: 10.1074/jbc.m116.765917] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 01/06/2017] [Indexed: 12/23/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), characterized by excessive fat accumulation in liver, is prevalent in obesity. Genetic factors that link obesity to NAFLD remain obscure. Apolipoprotein C3 (APOC3) is a lipid-binding protein with a pivotal role in triglyceride metabolism. Humans with APOC3 gain-of-function mutations and mice with APOC3 overproduction are associated with hypertriglyceridemia. Nonetheless, it remains controversial whether APOC3 is culpable for diet-induced NAFLD. To address this fundamental issue, we fed APOC3-transgenic and wild-type littermates a high fructose diet or high fat diet, followed by determination of the effect of APOC3 on hepatic lipid metabolism and inflammation and the progression of NAFLD. To gain mechanistic insight into NAFLD, we determined the impact of APOC3 on hepatic triglyceride synthesis and secretion versus fatty acid oxidation. APOC3-transgenic mice were hypertriglyceridemic, culminating in marked elevation of triglycerides, cholesterols, and non-esterified fatty acids in plasma. Despite the prevailing hypertriglyceridemia, APOC3-transgenic mice, relative to wild-type littermates, had similar weight gain and hepatic lipid content without alterations in hepatic expression of key genes involved in triglyceride synthesis and secretion and fatty acid oxidation. APOC3-transgenic and wild-type mice had similar Kupffer cell content without alterations in hepatic expression of pro- and anti-inflammatory cytokines. APOC3 neither exacerbated diet-induced adiposity nor aggravated the degree of steatosis in high fructose or high fat-fed APOC3-transgenic mice. These effects ensued independently of weight gain even after 10-month high fat feeding. We concluded that APOC3, whose dysregulation is liable for hypertriglyceridemia, is not a predisposing factor for linking overnutrition to NAFLD in obesity.
Collapse
Affiliation(s)
- Xiaoyun Cheng
- From the Department of Endocrinology and Metabolism, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai 200072, China and.,the Division of Endocrinology and Diabetes, Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| | - Jun Yamauchi
- the Division of Endocrinology and Diabetes, Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| | - Sojin Lee
- the Division of Endocrinology and Diabetes, Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| | - Ting Zhang
- the Division of Endocrinology and Diabetes, Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| | - Zhenwei Gong
- the Division of Endocrinology and Diabetes, Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| | - Radhika Muzumdar
- the Division of Endocrinology and Diabetes, Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| | - Shen Qu
- From the Department of Endocrinology and Metabolism, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai 200072, China and
| | - H Henry Dong
- the Division of Endocrinology and Diabetes, Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| |
Collapse
|
58
|
Erion DM, Park HJ, Lee HY. The role of lipids in the pathogenesis and treatment of type 2 diabetes and associated co-morbidities. BMB Rep 2017; 49:139-48. [PMID: 26728273 PMCID: PMC4915228 DOI: 10.5483/bmbrep.2016.49.3.268] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Indexed: 12/25/2022] Open
Abstract
In the past decade, the incidence of type 2 diabetes (T2D) has rapidly increased, along with the associated cardiovascular complications. Therefore, understanding the pathophysiology underlying T2D, the associated complications and the impact of therapeutics on the T2D development has critical importance for current and future therapeutics. The prevailing feature of T2D is hyperglycemia due to excessive hepatic glucose production, insulin resistance, and insufficient secretion of insulin by the pancreas. These contribute to increased fatty acid influx into the liver and muscle causing accumulation of lipid metabolites. These lipid metabolites cause dyslipidemia and non-alcoholic fatty liver disease, which ultimately contributes to the increased cardiovascular risk in T2D. Therefore, understanding the mechanisms of hepatic insulin resistance and the specific role of liver lipids is critical in selecting and designing the most effective therapeutics for T2D and the associated co-morbidities, including dyslipidemia and cardiovascular disease. Herein, we review the effects and molecular mechanisms of conventional anti-hyperglycemic and lipid-lowering drugs on glucose and lipid metabolism. [BMB Reports 2016; 49(3): 139-148].
Collapse
Affiliation(s)
- Derek M Erion
- Takeda Pharmaceuticals 350 Massachusetts Ave. Cambridge, MA, 02139, USA
| | - Hyun-Jun Park
- Department of Molecular Medicine, Lee Gil Ya Cancer and Diabetes Institute, School of Medicine, Gachon University, Incheon 21999, Korea
| | - Hui-Young Lee
- Department of Molecular Medicine and Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, School of Medicine, Gachon University, Incheon 21999, Korea
| |
Collapse
|
59
|
Apolipoprotein CIII Overexpression-Induced Hypertriglyceridemia Increases Nonalcoholic Fatty Liver Disease in Association with Inflammation and Cell Death. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:1838679. [PMID: 28163820 PMCID: PMC5259655 DOI: 10.1155/2017/1838679] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 10/26/2016] [Accepted: 11/24/2016] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the principal manifestation of liver disease in obesity and metabolic syndrome. By comparing hypertriglyceridemic transgenic mice expressing apolipoprotein (apo) CIII with control nontransgenic (NTg) littermates, we demonstrated that overexpression of apoCIII, independent of a high-fat diet (HFD), produces NAFLD-like features, including increased liver lipid content; decreased antioxidant power; increased expression of TNFα, TNFα receptor, cleaved caspase-1, and interleukin-1β; decreased expression of adiponectin receptor-2; and increased cell death. This phenotype is aggravated and additional NAFLD features are differentially induced in apoCIII mice fed a HFD. HFD induced glucose intolerance together with increased gluconeogenesis, indicating hepatic insulin resistance. Additionally, the HFD led to marked increases in plasma TNFα (8-fold) and IL-6 (60%) in apoCIII mice. Cell death signaling (Bax/Bcl2), effector (caspase-3), and apoptosis were augmented in apoCIII mice regardless of whether a HFD or a low-fat diet was provided. Fenofibrate treatment reversed several of the effects associated with diet and apoCIII expression but did not normalize inflammatory traits even when liver lipid content was fully corrected. These results indicate that apoCIII and/or hypertriglyceridemia plays a major role in liver inflammation and cell death, which in turn increases susceptibility to and the severity of diet-induced NAFLD.
Collapse
|
60
|
Abstract
The liver constitutes a key organ in systemic metabolism, contributing substantially to the development of insulin resistance and type 2 diabetes mellitus (T2DM). The mechanisms underlying these processes are not entirely understood, but involve hepatic fat accumulation, alterations of energy metabolism and inflammatory signals derived from various cell types including immune cells. Lipotoxins, mitochondrial function, cytokines and adipocytokines have been proposed to play a major part in both NAFLD and T2DM. Patients with NAFLD are commonly insulin resistant. On the other hand, a large number of patients with T2DM develop NAFLD with its inflammatory complication, NASH. The high incidence of NASH in patients with T2DM leads to further complications, such as liver cirrhosis and hepatocellular carcinoma, which are increasingly recognized. Therapeutic concepts such as thiazolidinediones (glitazones) for treating T2DM also show some efficacy in the treatment of NASH. This Review will describe the multifaceted and complex interactions between the liver and T2DM.
Collapse
|
61
|
Pesta DH, Perry RJ, Guebre-Egziabher F, Zhang D, Jurczak M, Fischer-Rosinsky A, Daniels MA, Willmes DM, Bhanot S, Bornstein SR, Knauf F, Samuel VT, Shulman GI, Birkenfeld AL. Prevention of diet-induced hepatic steatosis and hepatic insulin resistance by second generation antisense oligonucleotides targeted to the longevity gene mIndy (Slc13a5). Aging (Albany NY) 2016; 7:1086-93. [PMID: 26647160 PMCID: PMC4712334 DOI: 10.18632/aging.100854] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Reducing the expression of the Indy (I'm Not Dead Yet) gene in lower organisms extends life span by mechanisms resembling caloric restriction. Similarly, deletion of the mammalian homolog, mIndy (Slc13a5), encoding for a plasma membrane tricarboxylate transporter, protects from aging- and diet-induced adiposity and insulin resistance in mice. The organ specific contribution to this phenotype is unknown. We examined the impact of selective inducible hepatic knockdown of mIndy on whole body lipid and glucose metabolism using 2′-O-methoxyethyl chimeric anti-sense oligonucleotides (ASOs) in high-fat fed rats. 4-week treatment with 2′-O-methoxyethyl chimeric ASO reduced mIndy mRNA expression by 91% (P<0.001) compared to control ASO. Besides similar body weights between both groups, mIndy-ASO treatment lead to a 74% reduction in fasting plasma insulin concentrations as well as a 35% reduction in plasma triglycerides. Moreover, hepatic triglyceride content was significantly reduced by the knockdown of mIndy, likely mediating a trend to decreased basal rates of endogenous glucose production as well as an increased suppression of hepatic glucose production by 25% during a hyperinsulinemic-euglycemic clamp. Together, these data suggest that inducible liver-selective reduction of mIndy in rats is able to ameliorate hepatic steatosis and insulin resistance, conditions occurring with high calorie diets and during aging.
Collapse
Affiliation(s)
- Dominik H Pesta
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA.,Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA.,Department of Sport Science, Medical Section, University of Innsbruck, Innsbruck, Austria.,Department of Visceral, Transplant, and Thoracic Surgery, D. Swarovski Research Laboratory, Medical University of Innsbruck, Innsbruck, Austria.,Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University Düsseldorf, German Center for Diabetes Research, Partner Düsseldorf, Düsseldorf, Germany
| | - Rachel J Perry
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA.,Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA.,Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
| | | | - Dongyan Zhang
- Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Michael Jurczak
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Antje Fischer-Rosinsky
- Charité - University School of Medicine, Department of Endocrinology, Diabetes and Nutrition, Berlin, Germany
| | - Martin A Daniels
- Charité - University School of Medicine, Department of Endocrinology, Diabetes and Nutrition, Berlin, Germany.,Section of Metabolic Vascular Medicine, Medical Clinic III and Paul Langerhans Institute Dresden (PLID), TU Dresden, Germany
| | - Diana M Willmes
- Section of Metabolic Vascular Medicine, Medical Clinic III and Paul Langerhans Institute Dresden (PLID), TU Dresden, Germany.,German Center for Diabetes Research (DZD), Dresden, Germany
| | | | - Stefan R Bornstein
- Section of Metabolic Vascular Medicine, Medical Clinic III and Paul Langerhans Institute Dresden (PLID), TU Dresden, Germany.,German Center for Diabetes Research (DZD), Dresden, Germany.,Section of Diabetes and Nutritional Sciences, Rayne Institute, King's College London, London, UK
| | - Felix Knauf
- University Clinic Erlangen, Erlangen, Germany
| | - Varman T Samuel
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA.,Veterans Affairs Medical Center, West Haven, CT, USA
| | - Gerald I Shulman
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA.,Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA.,Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
| | - Andreas L Birkenfeld
- Section of Metabolic Vascular Medicine, Medical Clinic III and Paul Langerhans Institute Dresden (PLID), TU Dresden, Germany.,German Center for Diabetes Research (DZD), Dresden, Germany.,Section of Diabetes and Nutritional Sciences, Rayne Institute, King's College London, London, UK
| |
Collapse
|
62
|
Tao Y, Xiong Y, Wang H, Chu S, Zhong R, Wang J, Wang G, Ren X, Yu J. APOC3 induces endothelial dysfunction through TNF-α and JAM-1. Lipids Health Dis 2016; 15:153. [PMID: 27619170 PMCID: PMC5020557 DOI: 10.1186/s12944-016-0326-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Accepted: 09/06/2016] [Indexed: 01/13/2023] Open
Abstract
Background The fatality rate for cardiovascular disease (CVD) has increased in recent years and higher levels of triglyceride have been shown to be an independent risk factor for atherosclerotic CVD. Dysfunction of endothelial cells (ECs) is also a key factor of CVD. APOC3 is an important molecule in lipid metabolism that is closely associated with hyperlipidemia and an increased risk of developing CVD. But the direct effects of APOC3 on ECs were still unknown. This study was aimed at determining the effects of APOC3 on inflammation, chemotaxis and exudation in ECs. Methods ELISA, qRT-PCR, immunofluorescence, flow cytometry and transwell assays were used to investigate the effects of APOC3 on human umbilical vein endothelial cells (HUVECs). SiRNA-induced TNF-α and JAM-1 silencing were used to observe how APOC3 influenced the inflammatory process in the ECs. Results Our results showed that APOC3 was closely associated with the inflammatory process in ECs, and that this process was characterized by the increased expression of TNF-α. Inflammatory processes further disrupted the tight junctions (TJs) between HUVECs by causing increased expression of JAM-1. JAM-1 was involved in maintaining the integrity of TJs, and it promoted the assembly of platelets and the exudation of leukocytes. Changes in its expression promoted chemotaxis and the exudation of ECs, which contributed to atherosclerosis. While the integrity of the TJs was disrupted, the adhesion of THP-1 cells to HUVECs was also increased by APOC3. Conclusions In this study, we describe the mechanism by which APOC3 causes inflammation, chemotaxis and the exudation of ECs, and we suggest that controlling the inflammatory reactions that are caused by APOC3 may be a new method to treat CVD.
Collapse
Affiliation(s)
- Yun Tao
- Center of Laboratory Medicine, Affiliated Hospital, Nantong University, 20 Xi Si Road, Nantong, 226001, People's Republic of China
| | - Yisong Xiong
- Department of Laboratory Medicine, Chengdu Military General Hospital, 270 Tian Hui Road, Chengdu, 610000, People's Republic of China
| | - Huimin Wang
- Center of Laboratory Medicine, Affiliated Hospital, Nantong University, 20 Xi Si Road, Nantong, 226001, People's Republic of China
| | - Shaopeng Chu
- Center of Laboratory Medicine, Affiliated Hospital, Nantong University, 20 Xi Si Road, Nantong, 226001, People's Republic of China
| | - Renqian Zhong
- Department of Laboratory Medicine, Changzheng Hospital, Second Military Medical University, 415 Feng Yang Road, Shanghai, 200003, People's Republic of China
| | - Jianxin Wang
- Center of Laboratory Medicine, Affiliated Hospital, Nantong University, 20 Xi Si Road, Nantong, 226001, People's Republic of China
| | - Guihua Wang
- Center of Laboratory Medicine, Affiliated Hospital, Nantong University, 20 Xi Si Road, Nantong, 226001, People's Republic of China
| | - Xiumei Ren
- Center of Laboratory Medicine, Affiliated Hospital, Nantong University, 20 Xi Si Road, Nantong, 226001, People's Republic of China
| | - Juan Yu
- Center of Laboratory Medicine, Affiliated Hospital, Nantong University, 20 Xi Si Road, Nantong, 226001, People's Republic of China. .,Institute of Public Health, Nantong University, 9 Se Yuan Road, Nantong, 226001, People's Republic of China.
| |
Collapse
|
63
|
Capuani B, Pacifici F, Pastore D, Palmirotta R, Donadel G, Arriga R, Bellia A, Di Daniele N, Rogliani P, Abete P, Sbraccia P, Guadagni F, Lauro D, Della-Morte D. The role of epsilon PKC in acute and chronic diseases: Possible pharmacological implications of its modulators. Pharmacol Res 2016; 111:659-667. [PMID: 27461137 DOI: 10.1016/j.phrs.2016.07.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 07/22/2016] [Indexed: 02/06/2023]
|
64
|
Digenio A, Dunbar RL, Alexander VJ, Hompesch M, Morrow L, Lee RG, Graham MJ, Hughes SG, Yu R, Singleton W, Baker BF, Bhanot S, Crooke RM. Antisense-Mediated Lowering of Plasma Apolipoprotein C-III by Volanesorsen Improves Dyslipidemia and Insulin Sensitivity in Type 2 Diabetes. Diabetes Care 2016; 39:1408-15. [PMID: 27271183 DOI: 10.2337/dc16-0126] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 05/09/2016] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To determine the effects of volanesorsen (ISIS 304801), a second-generation 2'-O-methoxyethyl chimeric antisense inhibitor of apolipoprotein (apo)C-III, on triglyceride (TG) levels and insulin resistance in patients with type 2 diabetes. RESEARCH DESIGN AND METHODS A randomized, double-blind, placebo-controlled trial was performed in 15 adult patients with type 2 diabetes (HbA1c >7.5% [58 mmol/mol]) and hypertriglyceridemia (TG >200 and <500 mg/dL). Patients were randomized 2:1 to receive volanesorsen 300 mg or placebo for a total of 15 subcutaneous weekly doses. Glucose handling and insulin sensitivity were measured before and after treatment using a two-step hyperinsulinemic-euglycemic clamp procedure. RESULTS Treatment with volanesorsen significantly reduced plasma apoC-III (-88%, P = 0.02) and TG (-69%, P = 0.02) levels and raised HDL cholesterol (HDL-C) (42%, P = 0.03) compared with placebo. These changes were accompanied by a 57% improvement in whole-body insulin sensitivity (P < 0.001). Importantly, we found a strong relationship between enhanced insulin sensitivity and both plasma apoC-III (r = -0.61, P = 0.03) and TG (r = -0.68, P = 0.01) suppression. Improved insulin sensitivity was sufficient to significantly lower glycated albumin (-1.7%, P = 0.034) and fructosamine (-38.7 μmol/L, P = 0.045) at the end of dosing and HbA1c (-0.44% [-4.9 mmol/mol], P = 0.025) 3 months postdosing. CONCLUSIONS Volanesorsen reduced plasma apoC-III and TG while raising HDL-C levels. Importantly, glucose disposal, insulin sensitivity, and integrative markers of diabetes also improved in these patients after short-term treatment.
Collapse
Affiliation(s)
| | | | | | | | - Linda Morrow
- Profil Institute for Clinical Research, San Diego, CA
| | | | | | | | - Rosie Yu
- Ionis Pharmaceuticals, Inc., Carlsbad, CA
| | | | | | | | | |
Collapse
|
65
|
Ding Y, Xian X, Holland WL, Tsai S, Herz J. Low-Density Lipoprotein Receptor-Related Protein-1 Protects Against Hepatic Insulin Resistance and Hepatic Steatosis. EBioMedicine 2016; 7:135-45. [PMID: 27322467 PMCID: PMC4913705 DOI: 10.1016/j.ebiom.2016.04.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 03/31/2016] [Accepted: 04/01/2016] [Indexed: 11/15/2022] Open
Abstract
Low-density lipoprotein receptor-related protein-1 (LRP1) is a multifunctional uptake receptor for chylomicron remnants in the liver. In vascular smooth muscle cells LRP1 controls reverse cholesterol transport through platelet-derived growth factor receptor β (PDGFR-β) trafficking and tyrosine kinase activity. Here we show that LRP1 regulates hepatic energy homeostasis by integrating insulin signaling with lipid uptake and secretion. Somatic inactivation of LRP1 in the liver (hLRP1KO) predisposes to diet-induced insulin resistance with dyslipidemia and non-alcoholic hepatic steatosis. On a high-fat diet, hLRP1KO mice develop a severe Metabolic Syndrome secondary to hepatic insulin resistance, reduced expression of insulin receptors on the hepatocyte surface and decreased glucose transporter 2 (GLUT2) translocation. While LRP1 is also required for efficient cell surface insulin receptor expression in the absence of exogenous lipids, this latent state of insulin resistance is unmasked by exposure to fatty acids. This further impairs insulin receptor trafficking and results in increased hepatic lipogenesis, impaired fatty acid oxidation and reduced very low density lipoprotein (VLDL) triglyceride secretion. Hepatic LRP1 deficiency in a mouse model (hLRP1KO) predisposes to diet-induced insulin resistance, dyslipidemia, and obesity. Insulin resistance in the hLRP1KO mouse results from reduced cell surface expression of insulin receptor (IR) and impaired translocation of glucose transporter 2 (GLUT2). Excess fatty acids in hLRP1KO mice shift hepatic fatty acid metabolism from an oxidative to a synthetic state, resulting in hepatic steatosis.
LRP1 is a multifunctional transmembrane receptor with essential functions in lipoprotein metabolism and subcellular receptor tyrosine kinase trafficking. A mouse model of hepatic LRP1 deficiency integrates the hallmark findings in Metabolic Syndrome - insulin resistance, dyslipidemia, and hepatic steatosis - with impaired glucose metabolism and altered hepatic fatty acid metabolism as a consequence of reduced insulin receptor trafficking and signaling. These findings underscore the central role of LRP1 in overall energy homeostasis, and specifically liver glucose and fatty acid metabolism.
Collapse
Affiliation(s)
- Yinyuan Ding
- Department of Molecular Genetics, UT Southwestern Medical Center, Dallas, TX 75390, USA; Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX 75390, USA; Key Laboratory of Medical Electrophysiology, Ministry of Education of China, China; Institute of Cardiovascular Research, Sichuan Medical University, Luzhou 646000, China
| | - Xunde Xian
- Department of Molecular Genetics, UT Southwestern Medical Center, Dallas, TX 75390, USA; Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| | - William L Holland
- Department of Internal Medicine, Touchstone Diabetes Center, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Shirling Tsai
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA; Dallas VA Medical Center, Dallas, TX 75216, USA
| | - Joachim Herz
- Department of Molecular Genetics, UT Southwestern Medical Center, Dallas, TX 75390, USA; Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX 75390, USA; Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA; Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
66
|
Raposo HF, Paiva AA, Kato LS, de Oliveira HCF. Apolipoprotein CIII overexpression exacerbates diet-induced obesity due to adipose tissue higher exogenous lipid uptake and retention and lower lipolysis rates. Nutr Metab (Lond) 2015; 12:61. [PMID: 26705406 PMCID: PMC4690294 DOI: 10.1186/s12986-015-0058-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 10/29/2015] [Indexed: 12/15/2022] Open
Abstract
Background Hypertriglyceridemia is a common type of dyslipidemia found in obesity. However, it is not established whether primary hyperlipidemia can predispose to obesity. Evidences have suggested that proteins primarily related to plasma lipoprotein transport, such as apolipoprotein (apo) CIII and E, may significantly affect the process of body fat accumulation. We have previously observed an increased adiposity in response to a high fat diet (HFD) in mice overexpressing apoCIII. Here, we examined the potential mechanisms involved in this exacerbated response of apoCIII mice to the HFD. Methods We measured body energy balance, tissue capacity to store exogenous lipids, lipogenesis and lipolysis rates in non-transgenic and apoCIII overexpressing mice fed a HFD during two months. Results Food intake, fat excretion and whole body CO2 production were similar in both groups. However, the adipose tissue mass (45 %) and leptin plasma levels (2-fold) were significantly greater in apoCIII mice. Lipogenesis rates were similar, while exogenous lipid retention was increased in perigonadal (2-fold) and brown adipose tissues (40 %) of apoCIII mice. In addition, adipocyte basal lipolysis (55 %) and in vivo lipolysis index (30 %) were significantly decreased in apoCIII mice. A fat tolerance test evidenced delayed plasma triglyceride clearance and greater transient availability of non-esterified fatty acids (NEFA) during the post-prandial state in the apoCIII mice plasma. Thus, apoCIII overexpression resulted in increased NEFA availability to adipose uptake and decreased adipocyte lipolysis, favoring lipid enlargement of adipose depots. Conclusion We propose that plasma apoCIII levels represent a new risk factor for diet-induced obesity. Electronic supplementary material The online version of this article (doi:10.1186/s12986-015-0058-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Helena F Raposo
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas, SP Brazil
| | - Adriene A Paiva
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas, SP Brazil
| | - Larissa S Kato
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas, SP Brazil
| | - Helena C F de Oliveira
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas, SP Brazil ; Instituto de Biologia, Universidade Estadual de Campinas, Rua Monteiro Lobato, 255, Campinas, SP CEP 13083-862 Brazil
| |
Collapse
|
67
|
Asrih M, Jornayvaz FR. Metabolic syndrome and nonalcoholic fatty liver disease: Is insulin resistance the link? Mol Cell Endocrinol 2015; 418 Pt 1:55-65. [PMID: 25724480 DOI: 10.1016/j.mce.2015.02.018] [Citation(s) in RCA: 226] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 02/02/2015] [Accepted: 02/17/2015] [Indexed: 12/24/2022]
Abstract
Metabolic syndrome (MetS) is a disease composed of different risk factors such as obesity, type 2 diabetes or dyslipidemia. The prevalence of this syndrome is increasing worldwide in parallel with the rise in obesity. Nonalcoholic fatty liver disease (NAFLD) is now the most frequent chronic liver disease in western countries, affecting more than 30% of the general population. NAFLD encompasses a spectrum of liver manifestations ranging from simple steatosis to nonalcoholic steatohepatitis (NASH), fibrosis and cirrhosis, which may ultimately progress to hepatocellular carcinoma. There is accumulating evidence supporting an association between NAFLD and MetS. Indeed, NAFLD is recognized as the liver manifestation of MetS. Insulin resistance is increasingly recognized as a key factor linking MetS and NAFLD. Insulin resistance is associated with excessive fat accumulation in ectopic tissues, such as the liver, and increased circulating free fatty acids, which can further promote inflammation and endoplasmic reticulum stress. This in turn aggravates and maintains the insulin resistant state, constituting a vicious cycle. Importantly, evidence shows that most of the patients developing NAFLD present at least one of the MetS traits. This review will define MetS and NAFLD, provide an overview of the common pathophysiological mechanisms linking MetS and NAFLD, and give a perspective regarding treatment of these ever growing metabolic diseases.
Collapse
Affiliation(s)
- Mohamed Asrih
- Service of Endocrinology, Diabetes and Metabolism, Centre Hospitalier Universitaire Vaudois, Rue du Bugnon 46, Lausanne 1011, Switzerland
| | - François R Jornayvaz
- Service of Endocrinology, Diabetes and Metabolism, Centre Hospitalier Universitaire Vaudois, Rue du Bugnon 46, Lausanne 1011, Switzerland.
| |
Collapse
|
68
|
Norata GD, Tsimikas S, Pirillo A, Catapano AL. Apolipoprotein C-III: From Pathophysiology to Pharmacology. Trends Pharmacol Sci 2015; 36:675-687. [DOI: 10.1016/j.tips.2015.07.001] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 07/07/2015] [Accepted: 07/10/2015] [Indexed: 01/14/2023]
|
69
|
Apolipoprotein C3 Gene Variants and Risk of Developing Type 2 Diabetes in Saudi Subjects. Metab Syndr Relat Disord 2015; 13:298-303. [DOI: 10.1089/met.2015.0022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
70
|
Kalish BT, Fell GL, Nandivada P, Puder M. Clinically Relevant Mechanisms of Lipid Synthesis, Transport, and Storage. JPEN J Parenter Enteral Nutr 2015; 39:8S-17S. [PMID: 26187937 DOI: 10.1177/0148607115595974] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 05/26/2015] [Indexed: 12/19/2022]
Abstract
Lipids not only are fundamental nutrients but also serve as basic structural components of cells and as multifunctional signaling molecules. Lipid metabolism pathways underlie basic processes in health and disease and are the targets of novel therapeutics. In this review, we explore the molecular control of lipid synthesis, trafficking, and storage, with a focus on clinically relevant pathways. To illustrate the clinical relevance of molecular lipid regulation, we highlight how these biochemical processes contribute to the pathogenesis of nonalcoholic fatty liver disease, a component of the metabolic syndrome and a paradigmatic example of lipid dysregulation.
Collapse
Affiliation(s)
- Brian T Kalish
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Gillian L Fell
- Department of Surgery and The Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Prathima Nandivada
- Department of Surgery and The Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Mark Puder
- Department of Surgery and The Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
71
|
Berlanga A, Guiu-Jurado E, Porras JA, Aragonès G, Auguet T. [Role of metabolic lipases and lipotoxicity in the development of non-alcoholic steatosis and non-alcoholic steatohepatitis]. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS 2015; 28:47-61. [PMID: 26049666 DOI: 10.1016/j.arteri.2015.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 03/03/2015] [Accepted: 03/04/2015] [Indexed: 10/23/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become the most common liver disease in developed countries, covering a spectrum of pathological conditions ranging from single steatosis to non-alcoholic steatohepatitis, cirrhosis and hepatocellular carcinoma. Its pathogenesis has been often interpreted by the "double-hit" hypothesis, where the lipid accumulation in the liver is followed by proinflammatory mediators inducing inflammation, hepatocellular injury and fibrosis. Nowadays, a more complex model suggests that free fatty acids and their metabolites could be the true lipotoxic agents that contribute to the development of NAFLD and hepatic insulin resistance, suggesting a central role for metabolic lipases in that process.
Collapse
Affiliation(s)
- Alba Berlanga
- Grupo de recerca GEMMAIR (AGAUR)-Medicina Aplicada, Departamento de Medicina y Cirugía, Universidad Rovira i Virgili (URV), Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, España
| | - Esther Guiu-Jurado
- Grupo de recerca GEMMAIR (AGAUR)-Medicina Aplicada, Departamento de Medicina y Cirugía, Universidad Rovira i Virgili (URV), Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, España
| | - José Antonio Porras
- Grupo de recerca GEMMAIR (AGAUR)-Medicina Aplicada, Departamento de Medicina y Cirugía, Universidad Rovira i Virgili (URV), Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, España; Servicio de Medicina Interna, Hospital Universitario Joan XXIII, Tarragona, España
| | - Gemma Aragonès
- Grupo de recerca GEMMAIR (AGAUR)-Medicina Aplicada, Departamento de Medicina y Cirugía, Universidad Rovira i Virgili (URV), Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, España
| | - Teresa Auguet
- Grupo de recerca GEMMAIR (AGAUR)-Medicina Aplicada, Departamento de Medicina y Cirugía, Universidad Rovira i Virgili (URV), Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, España; Servicio de Medicina Interna, Hospital Universitario Joan XXIII, Tarragona, España.
| |
Collapse
|
72
|
Bell TA, Graham MJ, Baker BF, Crooke RM. Therapeutic inhibition of apoC-III for the treatment of hypertriglyceridemia. ACTA ACUST UNITED AC 2015. [DOI: 10.2217/clp.15.7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
73
|
Perry RJ, Zhang D, Zhang XM, Boyer JL, Shulman GI. Controlled-release mitochondrial protonophore reverses diabetes and steatohepatitis in rats. Science 2015; 347:1253-6. [PMID: 25721504 DOI: 10.1126/science.aaa0672] [Citation(s) in RCA: 214] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a major factor in the pathogenesis of type 2 diabetes (T2D) and nonalcoholic steatohepatitis (NASH). The mitochondrial protonophore 2,4 dinitrophenol (DNP) has beneficial effects on NAFLD, insulin resistance, and obesity in preclinical models but is too toxic for clinical use. We developed a controlled-release oral formulation of DNP, called CRMP (controlled-release mitochondrial protonophore), that produces mild hepatic mitochondrial uncoupling. In rat models, CRMP reduced hypertriglyceridemia, insulin resistance, hepatic steatosis, and diabetes. It also normalized plasma transaminase concentrations, ameliorated liver fibrosis, and improved hepatic protein synthetic function in a methionine/choline-deficient rat model of NASH. Chronic treatment with CRMP was not associated with any systemic toxicity. These data offer proof of concept that mild hepatic mitochondrial uncoupling may be a safe and effective therapy for the related epidemics of metabolic syndrome, T2D, and NASH.
Collapse
Affiliation(s)
- Rachel J Perry
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA. Departments of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA. Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Dongyan Zhang
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
| | - Xian-Man Zhang
- Departments of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - James L Boyer
- Departments of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA. Yale Liver Center, Yale University School of Medicine, New Haven, CT, USA
| | - Gerald I Shulman
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA. Departments of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA. Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
74
|
Camporez JPG, Kanda S, Petersen MC, Jornayvaz FR, Samuel VT, Bhanot S, Petersen KF, Jurczak MJ, Shulman GI. ApoA5 knockdown improves whole-body insulin sensitivity in high-fat-fed mice by reducing ectopic lipid content. J Lipid Res 2014; 56:526-536. [PMID: 25548259 PMCID: PMC4340301 DOI: 10.1194/jlr.m054080] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
ApoA5 has a critical role in the regulation of plasma TG concentrations. In order to determine whether ApoA5 also impacts ectopic lipid deposition in liver and skeletal muscle, as well as tissue insulin sensitivity, we treated mice with an antisense oligonucleotide (ASO) to decrease hepatic expression of ApoA5. ASO treatment reduced ApoA5 protein expression in liver by 60–70%. ApoA5 ASO-treated mice displayed approximately 3-fold higher plasma TG concentrations, which were associated with decreased plasma TG clearance. Furthermore, ApoA5 ASO-treated mice fed a high-fat diet (HFD) exhibited reduced liver and skeletal muscle TG uptake and reduced liver and muscle TG and diacylglycerol (DAG) content. HFD-fed ApoA5 ASO-treated mice were protected from HFD-induced insulin resistance, as assessed by hyperinsulinemic-euglycemic clamps. This protection could be attributed to increases in both hepatic and peripheral insulin responsiveness associated with decreased DAG activation of protein kinase C (PKC)-ε and PKCθ in liver and muscle, respectively, and increased insulin-stimulated AKT2 phosphorylation in these tissues. In summary, these studies demonstrate a novel role for ApoA5 as a modulator of susceptibility to diet-induced liver and muscle insulin resistance through regulation of ectopic lipid accumulation in liver and skeletal muscle.
Collapse
Affiliation(s)
| | - Shoichi Kanda
- Departments of Internal Medicine Yale University School of Medicine, New Haven, CT
| | - Max C Petersen
- Departments of Internal Medicine Yale University School of Medicine, New Haven, CT; Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT
| | - François R Jornayvaz
- Departments of Internal Medicine Yale University School of Medicine, New Haven, CT
| | - Varman T Samuel
- Departments of Internal Medicine Yale University School of Medicine, New Haven, CT
| | | | - Kitt Falk Petersen
- Departments of Internal Medicine Yale University School of Medicine, New Haven, CT
| | - Michael J Jurczak
- Departments of Internal Medicine Yale University School of Medicine, New Haven, CT
| | - Gerald I Shulman
- Departments of Internal Medicine Yale University School of Medicine, New Haven, CT; Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT; Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT.
| |
Collapse
|
75
|
Koppe SWP. Obesity and the liver: nonalcoholic fatty liver disease. Transl Res 2014; 164:312-22. [PMID: 25028077 DOI: 10.1016/j.trsl.2014.06.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 06/18/2014] [Accepted: 06/19/2014] [Indexed: 02/08/2023]
Abstract
The increasing prevalence of nonalcoholic fatty liver disease (NAFLD) parallels the rise of obesity and its complications. NAFLD is a common cause of cirrhosis and a leading indication for liver transplant. Genetic susceptibility, dietary composition, and exercise habits influence the development of NAFLD, and insulin resistance results in widespread metabolic perturbations with a net effect of triglyceride accumulation in the liver. Some patients will develop hepatocyte cellular injury and fibrosis of the liver, which can progress to cirrhosis and require liver transplant. Treatments targeting the pathophysiological mechanisms of NAFLD exist, but carry some potential risk and are not universally effective. Weight loss and lifestyle changes remain the most effective and safest approach, but sustainable change is difficult for most patients to achieve. Future work will continue to focus on developing effective and safe interventions to prevent the development of advanced liver disease, whereas efforts in the public health domain continue to combat obesity.
Collapse
Affiliation(s)
- Sean W P Koppe
- Division of Gastroenterology and Hepatology, Northwestern University, Chicago, Ill.
| |
Collapse
|
76
|
Affiliation(s)
- Gerald I Shulman
- From the Howard Hughes Medical Institute and the Departments of Internal Medicine and Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
77
|
Ehx G, Gérin S, Mathy G, Franck F, Oliveira HC, Vercesi AE, Sluse FE. Liver proteomic response to hypertriglyceridemia in human-apolipoprotein C-III transgenic mice at cellular and mitochondrial compartment levels. Lipids Health Dis 2014; 13:116. [PMID: 25047818 PMCID: PMC4112841 DOI: 10.1186/1476-511x-13-116] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 07/15/2014] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Hypertriglyceridemia (HTG) is defined as a triglyceride (TG) plasma level exceeding 150 mg/dl and is tightly associated with atherosclerosis, metabolic syndrome, obesity, diabetes and acute pancreatitis. The present study was undertaken to investigate the mitochondrial, sub-mitochondrial and cellular proteomic impact of hypertriglyceridemia in the hepatocytes of hypertriglyceridemic transgenic mice (overexpressing the human apolipoproteinC-III). METHODS Quantitative proteomics (2D-DIGE) analysis was carried out on both "low-expressor" (LE) and "high-expressor" (HE) mice, respectively exhibiting moderate and severe HTG, to characterize the effect of the TG plasma level on the proteomic response. RESULTS The mitoproteome analysis has revealed a large-scale phenomenon in transgenic mice, i.e. a general down-regulation of matricial proteins and up-regulation of inner membrane proteins. These data also demonstrate that the magnitude of proteomic changes strongly depends on the TG plasma level. Our different analyses indicate that, in HE mice, the capacity of several metabolic pathways is altered to promote the availability of acetyl-CoA, glycerol-3-phosphate, ATP and NADPH for TG de novo biosynthesis. The up-regulation of several cytosolic ROS detoxifying enzymes has also been observed, suggesting that the cytoplasm of HTG mice is subjected to oxidative stress. Moreover, our results suggest that iron over-accumulation takes place in the cytosol of HE mice hepatocytes and may contribute to enhance oxidative stress and to promote cellular proliferation. CONCLUSIONS These results indicate that the metabolic response to HTG in human apolipoprotein C-III overexpressing mice may support a high TG production rate and that the cytosol of hepatocytes is subjected to an important oxidative stress, probably as a result of FFA over-accumulation, iron overload and enhanced activity of some ROS-producing catabolic enzymes.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Francis E Sluse
- Laboratory of Bioenergetics (B22), Department of Life Sciences, University of Liege, Boulevard du rectorat 27, 4000 Liege, Belgium.
| |
Collapse
|
78
|
The role of hepatic lipids in hepatic insulin resistance and type 2 diabetes. Nature 2014; 510:84-91. [PMID: 24899308 DOI: 10.1038/nature13478] [Citation(s) in RCA: 822] [Impact Index Per Article: 82.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 03/14/2014] [Indexed: 02/07/2023]
Abstract
Non-alcoholic fatty liver disease and its downstream sequelae, hepatic insulin resistance and type 2 diabetes, are rapidly growing epidemics, which lead to increased morbidity and mortality rates, and soaring health-care costs. Developing interventions requires a comprehensive understanding of the mechanisms by which excess hepatic lipid develops and causes hepatic insulin resistance and type 2 diabetes. Proposed mechanisms implicate various lipid species, inflammatory signalling and other cellular modifications. Studies in mice and humans have elucidated a key role for hepatic diacylglycerol activation of protein kinase Cε in triggering hepatic insulin resistance. Therapeutic approaches based on this mechanism could alleviate the related epidemics of non-alcoholic fatty liver disease and type 2 diabetes.
Collapse
|
79
|
In-depth metabolic phenotyping of genetically engineered mouse models in obesity and diabetes. Mamm Genome 2014; 25:508-21. [PMID: 24792749 DOI: 10.1007/s00335-014-9520-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 04/10/2014] [Indexed: 01/09/2023]
Abstract
The world-wide prevalence of obesity and diabetes has increased sharply during the last two decades. Accordingly, the metabolic phenotyping of genetically engineered mouse models is critical for evaluating the functional roles of target genes in obesity and diabetes, and for developing new therapeutic targets. In this review, we discuss the practical meaning of metabolic phenotyping, the strategy of choosing appropriate tests, and considerations when designing and performing metabolic phenotyping in mice.
Collapse
|
80
|
Extrakorporale Therapien bei Patienten mit Lebererkrankungen auf der Intensivstation. Med Klin Intensivmed Notfmed 2014; 109:246-51. [DOI: 10.1007/s00063-013-0321-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 03/21/2014] [Indexed: 12/14/2022]
|
81
|
Asrih M, Jornayvaz FR. Diets and nonalcoholic fatty liver disease: The good and the bad. Clin Nutr 2014; 33:186-90. [DOI: 10.1016/j.clnu.2013.11.003] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 09/27/2013] [Accepted: 11/02/2013] [Indexed: 12/19/2022]
|
82
|
Fisher E, Lake E, McLeod RS. Apolipoprotein B100 quality control and the regulation of hepatic very low density lipoprotein secretion. J Biomed Res 2014; 28:178-93. [PMID: 25013401 PMCID: PMC4085555 DOI: 10.7555/jbr.28.20140019] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 02/15/2014] [Indexed: 12/19/2022] Open
Abstract
Apolipoprotein B (apoB) is the main protein component of very low density lipoprotein (VLDL) and is necessary for the assembly and secretion of these triglyceride (TG)-rich particles. Following release from the liver, VLDL is converted to low density lipoprotein (LDL) in the plasma and increased production of VLDL can therefore play a detrimental role in cardiovascular disease. Increasing evidence has helped to establish VLDL assembly as a target for the treatment of dyslipidemias. Multiple factors are involved in the folding of the apoB protein and the formation of a secretion-competent VLDL particle. Failed VLDL assembly can initiate quality control mechanisms in the hepatocyte that target apoB for degradation. ApoB is a substrate for endoplasmic reticulum associated degradation (ERAD) by the ubiquitin proteasome system and for autophagy. Efficient targeting and disposal of apoB is a regulated process that modulates VLDL secretion and partitioning of TG. Emerging evidence suggests that significant overlap exists between these degradative pathways. For example, the insulin-mediated targeting of apoB to autophagy and postprandial activation of the unfolded protein response (UPR) may employ the same cellular machinery and regulatory cues. Changes in the quality control mechanisms for apoB impact hepatic physiology and pathology states, including insulin resistance and fatty liver. Insulin signaling, lipid metabolism and the hepatic UPR may impact VLDL production, particularly during the postprandial state. In this review we summarize our current understanding of VLDL assembly, apoB degradation, quality control mechanisms and the role of these processes in liver physiology and in pathologic states.
Collapse
Affiliation(s)
- Eric Fisher
- Biochemistry & Molecular Biology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Elizabeth Lake
- Biochemistry & Molecular Biology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Roger S McLeod
- Biochemistry & Molecular Biology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
83
|
Birkenfeld AL, Shulman GI. Nonalcoholic fatty liver disease, hepatic insulin resistance, and type 2 diabetes. Hepatology 2014; 59:713-23. [PMID: 23929732 PMCID: PMC3946772 DOI: 10.1002/hep.26672] [Citation(s) in RCA: 522] [Impact Index Per Article: 52.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 07/31/2013] [Indexed: 12/12/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD), hepatic insulin resistance, and type 2 diabetes are all strongly associated and are all reaching epidemic proportions. Whether there is a causal link between NAFLD and hepatic insulin resistance is controversial. This review will discuss recent studies in both humans and animal models of NAFLD that have implicated increases in hepatic diacylglycerol (DAG) content leading to activation of novel protein kinase Cϵ (PKCϵ) resulting in decreased insulin signaling in the pathogenesis of NAFLD-associated hepatic insulin resistance and type 2 diabetes. The DAG-PKCϵ hypothesis can explain the occurrence of hepatic insulin resistance observed in most cases of NAFLD associated with obesity, lipodystrophy, and type 2 diabetes.
Collapse
Affiliation(s)
- Andreas L. Birkenfeld
- Charité - University School of Medicine, Department of Endocrinology Diabetes and Nutrition, Center for Cardiovascular Research, Berlin, Germany
- Howard Hughes Medical Institute and the Departments of Internal Medicine and Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Gerald I. Shulman
- Howard Hughes Medical Institute and the Departments of Internal Medicine and Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
84
|
Willmes DM, Birkenfeld AL. The Role of INDY in Metabolic Regulation. Comput Struct Biotechnol J 2013; 6:e201303020. [PMID: 24688728 PMCID: PMC3962103 DOI: 10.5936/csbj.201303020] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 12/02/2013] [Accepted: 12/02/2013] [Indexed: 01/20/2023] Open
Abstract
Reduced expression of the Indy (I'm Not Dead Yet) gene in D. melanogaster and C. elegans extends longevity. Indy and its mammalian homolog mINDY (Slc13a5, NaCT) are transporters of TCA cycle intermediates, mainly handling the uptake of citrate via the plasma membrane into the cytosol. Deletion of mINDY in mice leads to significant metabolic changes akin to caloric restriction, likely caused by reducing the effects of mINDY-imported citrate on fatty acid and cholesterol synthesis, glucose metabolism and ß-oxidation. This review will provide an overview on different mammalian SLC1 3 family members with a focus on mINDY (SLCl3A5) in glucose and energy metabolism and will highlight the role of mINDY as a putative therapeutic target for the treatment of obesity, non-alcoholic fatty liver disease and type 2 diabetes.
Collapse
Affiliation(s)
- Diana M Willmes
- Department of Endocrinology, Diabetes and Nutrition, Center for Cardiovascular Research, Charité - University School of Medicine, Berlin, Germany
| | - Andreas L Birkenfeld
- Department of Endocrinology, Diabetes and Nutrition, Center for Cardiovascular Research, Charité - University School of Medicine, Berlin, Germany
| |
Collapse
|
85
|
Walker RW, Sinatra F, Hartiala J, Weigensberg M, Spruijt-Metz D, Alderete TL, Goran MI, Allayee H. Genetic and clinical markers of elevated liver fat content in overweight and obese Hispanic children. Obesity (Silver Spring) 2013; 21:E790-7. [PMID: 23804528 PMCID: PMC3855210 DOI: 10.1002/oby.20523] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 05/29/2013] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Genetic variation in six genes has been associated with elevated liver fat and nonalcoholic fatty liver disease in adults. The influence of these genes on liver fat and whether a genetic risk score (GRS) would improve upon the ability of common clinical risk factors to predict elevated liver fat content (ELF) in Hispanic children was determined. DESIGN AND METHODS 223 obese Hispanic children were genotyped for six SNPs. MRI was used to measure liver fat. A GRS was tested for association with ELF using multivariate linear regression. Predictors were assessed via ROC curves and pair-wise analysis was used to determine significance alone and combined with clinical markers. RESULTS Only variants in PNPLA3 and APOC3 genes were associated with liver fat (P < 0.001, P = 0.01, respectively). Subjects with a GRS = 4 had ∼3-fold higher liver fat content than subjects with GRS of 0 (15.1 ± 12.7 vs. 5.1 ± 3.7%, P = 0.03). While the addition of the GRS to a model containing BMI and liver enzymes increased ROC AUC from 0.83 to 0.85 [95% CI, 0.79-0.89], (P = 0.01), it does not improve detection of ELF from a clinical perspective. CONCLUSIONS Only PNPLA3 and APOC3 were related to ELF and a GRS comprised of these susceptibility alleles did not add to the discriminatory power of traditional biomarkers for clinical assessment of liver fat.
Collapse
Affiliation(s)
- Ryan W Walker
- Department of Preventive Medicine, University of Southern California, Keck School of Medicine, Los Angeles, USA
| | | | | | | | | | | | | | | |
Collapse
|
86
|
Perry RJ, Kim T, Zhang XM, Lee HY, Pesta D, Popov VB, Zhang D, Rahimi Y, Jurczak MJ, Cline GW, Spiegel DA, Shulman GI. Reversal of hypertriglyceridemia, fatty liver disease, and insulin resistance by a liver-targeted mitochondrial uncoupler. Cell Metab 2013; 18:740-8. [PMID: 24206666 PMCID: PMC4104686 DOI: 10.1016/j.cmet.2013.10.004] [Citation(s) in RCA: 179] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Revised: 08/12/2013] [Accepted: 10/04/2013] [Indexed: 12/22/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) affects one in three Americans and is a major predisposing condition for the metabolic syndrome and type 2 diabetes (T2D). We examined whether a functionally liver-targeted derivative of 2,4-dinitrophenol (DNP), DNP-methyl ether (DNPME), could safely decrease hypertriglyceridemia, NAFLD, and insulin resistance without systemic toxicities. Treatment with DNPME reversed hypertriglyceridemia, fatty liver, and whole-body insulin resistance in high-fat-fed rats and decreased hyperglycemia in a rat model of T2D with a wide therapeutic index. The reversal of liver and muscle insulin resistance was associated with reductions in tissue diacylglycerol content and reductions in protein kinase C epsilon (PKCε) and PKCθ activity in liver and muscle, respectively. These results demonstrate that the beneficial effects of DNP on hypertriglyceridemia, fatty liver, and insulin resistance can be dissociated from systemic toxicities and suggest the potential utility of liver-targeted mitochondrial uncoupling agents for the treatment of hypertriglyceridemia, NAFLD, metabolic syndrome, and T2D.
Collapse
Affiliation(s)
- Rachel J. Perry
- Howard Hughes Medical Institute, Yale University School of Medicine New Haven, CT, USA 06519
- Department of Internal Medicine, Yale University School of Medicine New Haven, CT, USA 06519
- Department of Cellular & Molecular Physiology, Yale University School of Medicine New Haven, CT, USA 06519
| | - Taehan Kim
- Department of Pharmacology, Yale University School of Medicine New Haven, CT, USA 06519
| | - Xian-Man Zhang
- Howard Hughes Medical Institute, Yale University School of Medicine New Haven, CT, USA 06519
| | - Hui-Young Lee
- Howard Hughes Medical Institute, Yale University School of Medicine New Haven, CT, USA 06519
- Department of Cellular & Molecular Physiology, Yale University School of Medicine New Haven, CT, USA 06519
| | - Dominik Pesta
- Howard Hughes Medical Institute, Yale University School of Medicine New Haven, CT, USA 06519
| | - Violeta B. Popov
- Department of Internal Medicine, Yale University School of Medicine New Haven, CT, USA 06519
| | - Dongyan Zhang
- Howard Hughes Medical Institute, Yale University School of Medicine New Haven, CT, USA 06519
| | - Yasmeen Rahimi
- Howard Hughes Medical Institute, Yale University School of Medicine New Haven, CT, USA 06519
| | - Michael J. Jurczak
- Howard Hughes Medical Institute, Yale University School of Medicine New Haven, CT, USA 06519
| | - Gary W. Cline
- Howard Hughes Medical Institute, Yale University School of Medicine New Haven, CT, USA 06519
| | - David A. Spiegel
- Department of Pharmacology, Yale University School of Medicine New Haven, CT, USA 06519
- Department of Chemistry, Yale University, New Haven, CT, USA 06520
| | - Gerald I. Shulman
- Howard Hughes Medical Institute, Yale University School of Medicine New Haven, CT, USA 06519
- Department of Internal Medicine, Yale University School of Medicine New Haven, CT, USA 06519
- Department of Cellular & Molecular Physiology, Yale University School of Medicine New Haven, CT, USA 06519
- Novo Nordisk Foundation Center for Basic Biomedical Research Copenhagen, DK
- Correspondence to:
| |
Collapse
|
87
|
Camporez JPG, Jornayvaz FR, Petersen MC, Pesta D, Guigni BA, Serr J, Zhang D, Kahn M, Samuel VT, Jurczak MJ, Shulman GI. Cellular mechanisms by which FGF21 improves insulin sensitivity in male mice. Endocrinology 2013; 154:3099-109. [PMID: 23766126 PMCID: PMC3749479 DOI: 10.1210/en.2013-1191] [Citation(s) in RCA: 167] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Fibroblast growth factor 21 (FGF21) is a potent regulator of glucose and lipid metabolism and is currently being pursued as a therapeutic agent for insulin resistance and type 2 diabetes. However, the cellular mechanisms by which FGF21 modifies insulin action in vivo are unclear. To address this question, we assessed insulin action in regular chow- and high-fat diet (HFD)-fed wild-type mice chronically infused with FGF21 or vehicle. Here, we show that FGF21 administration results in improvements in both hepatic and peripheral insulin sensitivity in both regular chow- and HFD-fed mice. This improvement in insulin responsiveness in FGF21-treated HFD-fed mice was associated with decreased hepatocellular and myocellular diacylglycerol content and reduced protein kinase Cε activation in liver and protein kinase Cθ in skeletal muscle. In contrast, there were no effects of FGF21 on liver or muscle ceramide content. These effects may be attributed, in part, to increased energy expenditure in the liver and white adipose tissue. Taken together, these data provide a mechanism by which FGF21 protects mice from lipid-induced liver and muscle insulin resistance and support its development as a novel therapy for the treatment of nonalcoholic fatty liver disease, insulin resistance, and type 2 diabetes.
Collapse
MESH Headings
- Adipose Tissue, Brown/drug effects
- Adipose Tissue, Brown/metabolism
- Adipose Tissue, Brown/surgery
- Animals
- Cells, Cultured
- Diet, High-Fat/adverse effects
- Drug Implants
- Energy Metabolism/drug effects
- Fibroblast Growth Factors/administration & dosage
- Fibroblast Growth Factors/metabolism
- Fibroblast Growth Factors/therapeutic use
- Glucose Intolerance/drug therapy
- Glucose Intolerance/etiology
- Glucose Intolerance/metabolism
- Glucose Intolerance/pathology
- Humans
- Infusions, Subcutaneous
- Insulin Resistance
- Isoenzymes/metabolism
- Lipectomy
- Lipid Metabolism/drug effects
- Liver/drug effects
- Liver/metabolism
- Liver/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Protein Kinase C/metabolism
- Protein Kinase C-epsilon/metabolism
- Protein Kinase C-theta
- Recombinant Proteins/administration & dosage
- Recombinant Proteins/metabolism
- Recombinant Proteins/therapeutic use
Collapse
Affiliation(s)
- João Paulo G Camporez
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06536-9812, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Asrih M, Jornayvaz FR. Inflammation as a potential link between nonalcoholic fatty liver disease and insulin resistance. J Endocrinol 2013; 218:R25-36. [PMID: 23833274 DOI: 10.1530/joe-13-0201] [Citation(s) in RCA: 208] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has become a major health problem in developed countries. It has affected more than 30% of the general population and is commonly associated with insulin resistance, which is a major risk factor for the development of type 2 diabetes and a central feature of the metabolic syndrome. Furthermore, accumulating evidences reveal that NAFLD as well as insulin resistance is strongly related to inflammation. Cytokines and adipokines play a pivotal role in inflammatory processes. In addition, these inflammatory mediators regulate various functions including metabolic energy balance, inflammation, and immune response. However, their role in modulating ectopic lipids involved in the development of insulin resistance, such as diacylglycerols and ceramides, remains unknown. The aim of this review is first to describe the pathophysiology of insulin resistance in NAFLD. In particular, we discuss the role of ectopic lipid accumulation in the liver. Secondly, we also summarize recent findings emphasizing the role of main inflammatory markers in both NAFLD and insulin resistance and their potential role in modulating hepatic fat content in NAFLD and associated hepatic insulin resistance.
Collapse
Affiliation(s)
- Mohamed Asrih
- Service of Endocrinology, Diabetes, Hypertension and Nutrition, Geneva University Hospital, Rue Gabrielle-Perret-Gentil 4, 1211 Genève 14, Switzerland
| | | |
Collapse
|
89
|
Zampino R, Coppola N, Cirillo G, Boemio A, Pisaturo M, Marrone A, Macera M, Sagnelli E, Perrone L, Adinolfi LE, Miraglia del Giudice E. Abdominal fat interacts with PNPLA3 I148M, but not with the APOC3 variant in the pathogenesis of liver steatosis in chronic hepatitis C. J Viral Hepat 2013; 20:517-23. [PMID: 23808989 DOI: 10.1111/jvh.12053] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 12/02/2012] [Indexed: 12/12/2022]
Abstract
The patatin-like phospholipase domain-containing 3 gene (PNPLA3) and the apolipoprotein C3 gene (APOC3) have been studied in relation to liver steatosis and liver disease outcome. The aim of this study was to evaluate the influence of PNPLA3 p.I148M and APOC3 rs2854116 and rs2854117 polymorphisms on the clinical and histological presentation of chronic hepatitis C in an Italian population and their relationship with viral and anthropometric parameters. Patients with hepatitis C (n = 166) entered the study receiving a clinical, histological, virological and biochemical evaluation. APOC3 (rs2854116 and rs2854117) and PNPLA3 (p.I148M) variants were genotyped. PNPLA3 polymorphisms were associated with liver steatosis, which was significantly higher in patients with p.148I/M (P = 0.034) and p.148M/M (P = 0.004) variants than those homozygous for the PNPLA3 wild type. Excluding patients with HCV genotype 3, the association with liver steatosis and PNPLA3 variants was more marked (p.148I/I genotype vs p.148I/M, P = 0.02, and vs p.148M/M, P = 0.005). The APOC3 polymorphism was not associated with any of the evaluated parameters. Among the interacting factors, BMI and waist circumference correlated with liver steatosis (P = 0.008 and 0.004, respectively). Relationship between waist circumference and liver steatosis was analysed for the different PNPLA3 genotypes. Homozygous 148M patients showed a stronger correlation between waist circumference and steatosis than those carrying the other genotypes (P = 0.0047). In our hepatitis C-infected population, the PNPLA3 polymorphism influenced the development of liver steatosis, but not fibrosis progression. APOC3 polymorphisms had no effect on the development of steatosis and no influence on the PNPLA3 polymorphism. The amount of abdominal fat can increase the association of PNPLA3 p.I148M with liver steatosis.
Collapse
Affiliation(s)
- R Zampino
- Internal Medicine and Hepatology, Second University of Naples, Naples, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Lee RG, Crosby J, Baker BF, Graham MJ, Crooke RM. Antisense technology: an emerging platform for cardiovascular disease therapeutics. J Cardiovasc Transl Res 2013; 6:969-80. [PMID: 23856914 PMCID: PMC3838598 DOI: 10.1007/s12265-013-9495-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 06/21/2013] [Indexed: 12/25/2022]
Abstract
Antisense oligonucleotides and small interfering RNAs, which suppress the translation of specific mRNA target proteins, are emerging as important therapeutic modalities for the treatment of cardiovascular disease. Over the last 25 years, the advances in all aspects of antisense technology, as well as a detailed understanding of the mechanism of action of antisense drugs, have enabled their use as therapeutic agents. These advancements culminated in the FDA approval of the first chronically administered cardiovascular antisense therapeutic, mipomersen, which targets hepatic apolipoprotein B mRNA. This review provides a brief history of antisense technology, highlights the progression of mipomersen from preclinical studies to multiple Phase III registration trials, and gives an update on the status of other cardiovascular antisense therapeutics currently in the clinic.
Collapse
Affiliation(s)
- Richard G Lee
- Isis Pharmaceuticals, Inc., 2855 Gazelle Court, Carlsbad, CA, 92010, USA,
| | | | | | | | | |
Collapse
|
91
|
Schmitz-Peiffer C. The tail wagging the dog--regulation of lipid metabolism by protein kinase C. FEBS J 2013; 280:5371-83. [PMID: 23587021 DOI: 10.1111/febs.12285] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2013] [Revised: 03/19/2013] [Accepted: 04/11/2013] [Indexed: 12/12/2022]
Abstract
Upon their discovery almost 40 years ago, isoforms of the lipid-activated protein kinase C (PKC) family were initially regarded only as downstream effectors of the second messengers calcium and diacylglycerol, undergoing activation upon phospholipid hydrolysis in response to acute stimuli. Subsequently, several isoforms were found to be associated with the inhibitory effects of lipid over-supply on glucose homeostasis, especially the negative cross-talk with insulin signal transduction, observed upon accumulation of diacylglycerol in insulin target tissues. The PKC family has therefore attracted much attention in diabetes and obesity research, because intracellular lipid accumulation is strongly correlated with defective insulin action and the development of type 2 diabetes. Causal roles for various isoforms in the generation of insulin resistance have more recently been confirmed using PKC-deficient mice. However, during characterization of these animals, it became increasingly evident that the enzymes play key roles in the modulation of lipid metabolism itself, and may control the supply of lipids between tissues such as adipose and liver. Molecular studies have also demonstrated roles for PKC isoforms in several aspects of lipid metabolism, such as adipocyte differentiation and hepatic lipogenesis. While the precise mechanisms involved, especially the identities of protein substrates, are still unclear, the emerging picture suggests that the currently held view of the contribution of PKC isoforms to metabolism is an over-simplification. Although PKCs may inhibit insulin signal transduction, these enzymes are not merely downstream effectors of lipid accumulation, but in fact control the fate of fatty acids, thus the tail wags the dog.
Collapse
Affiliation(s)
- Carsten Schmitz-Peiffer
- Diabetes and Obesity Program, Garvan Institute of Medical Research, Sydney, NSW, Australia; St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
92
|
Yao J, Zhi M, Gao X, Hu P, Li C, Yang X. Effect and the probable mechanisms of silibinin in regulating insulin resistance in the liver of rats with non-alcoholic fatty liver. Braz J Med Biol Res 2013; 46:270-7. [PMID: 23532271 PMCID: PMC3854368 DOI: 10.1590/1414-431x20122551] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Accepted: 12/04/2012] [Indexed: 02/07/2023] Open
Abstract
Our previous study has shown that reduced insulin resistance (IR) was one of the
possible mechanisms for the therapeutic effect of silibinin on non-alcoholic
fatty liver disease (NAFLD) in rats. In the present study, we investigated the
pathways of silibinin in regulating hepatic glucose production and IR
amelioration. Forty-five 4- to 6-week-old male Sprague Dawley rats were divided
into a control group, an HFD group (high-fat diet for 6 weeks) and an HFD +
silibinin group (high-fat diet + 0.5 mg kg-1·day-1
silibinin, starting at the beginning of the protocol). Both subcutaneous and
visceral fat was measured. Homeostasis model assessment-IR index (HOMA-IR),
intraperitoneal glucose tolerance test and insulin tolerance test (ITT) were
performed. The expression of adipose triglyceride lipase (ATGL) and of genes
associated with hepatic gluconeogenesis was evaluated. Silibinin intervention
significantly protected liver function, down-regulated serum fat, and improved
IR, as shown by decreased HOMA-IR and increased ITT slope. Silibinin markedly
prevented visceral obesity by reducing visceral fat, enhanced lipolysis by
up-regulating ATGL expression and inhibited gluconeogenesis by down-regulating
associated genes such as Forkhead box O1, phosphoenolpyruvate carboxykinase and
glucose-6-phosphatase. Silibinin was effective in ameliorating IR in NAFLD rats.
Reduction of visceral obesity, enhancement of lipolysis and inhibition of
gluconeogenesis might be the underlying mechanisms.
Collapse
Affiliation(s)
- Jiayin Yao
- Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, People’s Republic of China
| | | | | | | | | | | |
Collapse
|
93
|
Corbin KD, Abdelmalek MF, Spencer MD, da Costa KA, Galanko JA, Sha W, Suzuki A, Guy CD, Cardona DM, Torquati A, Diehl AM, Zeisel SH. Genetic signatures in choline and 1-carbon metabolism are associated with the severity of hepatic steatosis. FASEB J 2013; 27:1674-89. [PMID: 23292069 DOI: 10.1096/fj.12-219097] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Choline metabolism is important for very low-density lipoprotein secretion, making this nutritional pathway an important contributor to hepatic lipid balance. The purpose of this study was to assess whether the cumulative effects of multiple single nucleotide polymorphisms (SNPs) across genes of choline/1-carbon metabolism and functionally related pathways increase susceptibility to developing hepatic steatosis. In biopsy-characterized cases of nonalcoholic fatty liver disease and controls, we assessed 260 SNPs across 21 genes in choline/1-carbon metabolism. When SNPs were examined individually, using logistic regression, we only identified a single SNP (PNPLA3 rs738409) that was significantly associated with severity of hepatic steatosis after adjusting for confounders and multiple comparisons (P=0.02). However, when groupings of SNPs in similar metabolic pathways were defined using unsupervised hierarchical clustering, we identified groups of subjects with shared SNP signatures that were significantly correlated with steatosis burden (P=0.0002). The lowest and highest steatosis clusters could also be differentiated by ethnicity. However, unique SNP patterns defined steatosis burden irrespective of ethnicity. Our results suggest that analysis of SNP patterns in genes of choline/1-carbon metabolism may be useful for prediction of severity of steatosis in specific subsets of people, and the metabolic inefficiencies caused by these SNPs should be examined further.
Collapse
Affiliation(s)
- Karen D Corbin
- University of North Carolina at Chapel Hill Nutrition Research Institute, Kannapolis, NorthCarolina 28081, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Blackett PR, Sanghera DK. Genetic determinants of cardiometabolic risk: a proposed model for phenotype association and interaction. J Clin Lipidol 2013; 7:65-81. [PMID: 23351585 PMCID: PMC3559023 DOI: 10.1016/j.jacl.2012.04.079] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Revised: 03/16/2012] [Accepted: 04/16/2012] [Indexed: 12/15/2022]
Abstract
This review provides a translational and unifying summary of metabolic syndrome genetics and highlights evidence that genetic studies are starting to unravel and untangle origins of the complex and challenging cluster of disease phenotypes. The associated genes effectively express in the brain, liver, kidney, arterial endothelium, adipocytes, myocytes, and β cells. Progression of syndrome traits has been associated with ectopic lipid accumulation in the arterial wall, visceral adipocytes, myocytes, and liver. Thus, it follows that the genetics of dyslipidemia, obesity, and nonalcoholic fatty liver disease are central in triggering progression of the syndrome to overt expression of disease traits and have become a key focus of interest for early detection and for designing prevention and treatments. To support the "birds' eye view" approach, we provide a road-map depicting commonality and interrelationships between the traits and their genetic and environmental determinants based on known risk factors, metabolic pathways, pharmacologic targets, treatment responses, gene networks, pleiotropy, and association with circadian rhythm. Although only a small portion of the known heritability is accounted for and there is insufficient support for clinical application of gene-based prediction models, there is direction and encouraging progress in a rapidly moving field that is beginning to show clinical relevance.
Collapse
Affiliation(s)
- Piers R Blackett
- Department of Pediatrics, 940 NE 13St., University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Dharambir K Sanghera
- Department of Pediatrics, 940 NE 13St., University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| |
Collapse
|
95
|
Gariani K, Philippe J, Jornayvaz FR. Non-alcoholic fatty liver disease and insulin resistance: from bench to bedside. DIABETES & METABOLISM 2012; 39:16-26. [PMID: 23266468 DOI: 10.1016/j.diabet.2012.11.002] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Revised: 11/09/2012] [Accepted: 11/10/2012] [Indexed: 02/07/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is now the most frequent chronic liver disease in the developed countries. There is also growing evidence from basic and clinical research that NAFLD has a strong relationship to insulin resistance, which is a key factor in the development of type 2 diabetes. The aim of this review is to summarize the recent important findings linking NAFLD and insulin resistance. Lipid accumulation, particularly of diacylglycerol, appears to be of major importance in this process. Mitochondrial dysfunction, through decreased mitochondrial biogenesis, increases oxidative stress, and ageing also plays an important role. Finally, endoplasmic reticulum stress and inflammation also probably contribute to the development of insulin resistance via mechanisms that are still not well understood. Clinical aspects of NAFLD, such as its diagnosis and management, are also investigated in this review.
Collapse
Affiliation(s)
- K Gariani
- Service of Endocrinology, Diabetes, Hypertension and Nutrition, Geneva University Hospitals, rue Gabrielle-Perret-Gentil 4, 1211 Geneva 14, Switzerland
| | | | | |
Collapse
|
96
|
Jiang ZG, Robson SC, Yao Z. Lipoprotein metabolism in nonalcoholic fatty liver disease. J Biomed Res 2012; 27:1-13. [PMID: 23554788 PMCID: PMC3596749 DOI: 10.7555/jbr.27.20120077] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Revised: 08/23/2012] [Accepted: 08/29/2012] [Indexed: 12/18/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), an escalating health problem worldwide, covers a spectrum of pathologies characterized by fatty accumulation in hepatocytes in early stages, with potential progression to liver inflammation, fibrosis, and failure. A close, yet poorly understood link exists between NAFLD and dyslipidemia, a constellation of abnormalities in plasma lipoproteins including triglyceride-rich very low density lipoproteins. Apolipoproteins are a group of primarily liver-derived proteins found in serum lipoproteins; they not only play an extracellular role in lipid transport between vital organs through circulation, but also play an important intracellular role in hepatic lipoprotein assembly and secretion. The liver functions as the central hub for lipoprotein metabolism, as it dictates lipoprotein production and to a significant extent modulates lipoprotein clearance. Lipoprotein metabolism is an integral component of hepatocellular lipid homeostasis and is implicated in the pathogenesis, potential diagnosis, and treatment of NAFLD.
Collapse
Affiliation(s)
- Zhenghui Gordon Jiang
- Department of Medicine, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA, USA
| | | | | |
Collapse
|
97
|
Abstract
PURPOSE OF REVIEW This article reviews the mechanisms leading to the development of nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH) and the effects of hypoglycaemic and lipid-lowering therapies on NAFLD/NASH. RECENT FINDINGS The interaction of lipogenesis, fatty acid oxidation, inflammation, endoplasmic reticulum stress and hepatic insulin resistance contribute to the pathogenesis of NAFLD/NASH. Few large scale clinical trials exist with biopsy or magnetic resonance endpoints as opposed to ultrasonographic and transaminase endpoints. Trial evidence that exists supports the utility of weight loss, metformin, thiazolidinediones, fibrates, niacin, ezetimibe and statins in improving the steatosis component of NAFLD/NASH though with less or minimal effects on the fibrotic component of NASH. SUMMARY Hypoglycaemic and lipid-lowering therapies may have a role in the treatment of NAFLD/NASH but large scale endpoint trials remain to be performed.
Collapse
Affiliation(s)
- Anthony S Wierzbicki
- Department of Metabolic Medicine/Chemical Pathologyemical Pathology, Guy's and St. Thomas' Hospitals, London, UK.
| | | |
Collapse
|
98
|
Torres DM, Williams CD, Harrison SA. Features, diagnosis, and treatment of nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol 2012; 10:837-58. [PMID: 22446927 DOI: 10.1016/j.cgh.2012.03.011] [Citation(s) in RCA: 182] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Accepted: 03/13/2012] [Indexed: 02/06/2023]
Abstract
As the global incidence of obesity has increased, nonalcoholic fatty liver disease (NAFLD) has become a worldwide health concern. NAFLD occurs in children and adults of all ethnicities and includes isolated fatty liver and nonalcoholic steatohepatitis (NASH). Patients with NASH are at risk for developing cirrhosis, hepatic decompensation, and hepatocellular carcinoma and have increased all-cause mortality. NAFLD is associated with a variety of clinical conditions and is an independent risk factor for hepatocellular carcinoma. The pathogenesis of NAFLD and the specific steps that lead to NASH and advanced fibrosis are not fully understood, although researchers have found that a combination of environmental, genetic, and metabolic factors lead to advanced disease. There have been improvements in noninvasive radiographic methods to diagnose NAFLD, especially for advanced disease. However, liver biopsy is still the standard method of diagnosis for NASH. There are many challenges to treating patients with NASH, and no therapies have been approved by the U.S. Food and Drug Administration; multimodal approaches are being developed and becoming the standard of care. We review pathogenesis and treatment approaches for the West's largest liver-related public health concern.
Collapse
Affiliation(s)
- Dawn M Torres
- Division of Gastroenterology, Department of Medicine, Walter Reed National Military Medical Center, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
99
|
Abstract
PURPOSE OF REVIEW A strong positive correlation between plasma apolipoprotein (apo) C-III and triglyceride concentrations has been invariably observed in human and animal studies. The hypertriglyceridemic effect of apo C-III has been conventionally explained by its extracellular roles in inhibiting lipolysis catalysed by lipoprotein lipase and attenuating triglyceride-rich lipoprotein clearance through receptor-dependent and/or independent mechanisms. However, recent experimental evidence suggests that apo C-III may also play an intracellular role in promoting hepatic triglyceride-rich lipoprotein production. RECENT FINDINGS Kinetic studies with humans and genetically modified mice have shown that apo C-III is linked with increased production of triglyceride-rich lipoproteins, such as very-low-density lipoprotein 1 (VLDL1). Mutational studies on human apo C-III variants (originally identified in humans with hypotriglyceridemia or hyperalphalipoproteinemia) provide the structure-function analysis of human apo C-III, demonstrating that loss-of-function mutations within human apo C-III impair the assembly and secretion of triglyceride-rich VLDL1 under lipid-rich conditions. SUMMARY The current review summarizes recent experimental evidence for an intrahepatic role of human apo C-III in promoting mobilization and utilization of triglyceride during VLDL1 assembly/secretion. Understanding mechanisms by which hepatic apo C-III expression is regulated under insulin resistance and diabetic conditions will lead to better and more rational strategies for the prevention and treatment of diabetic hypertriglyceridemia that is closely related to premature atherosclerosis.
Collapse
Affiliation(s)
- Zemin Yao
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| | | |
Collapse
|
100
|
Abstract
Nonalcoholic fatty liver disease (NAFLD) is now the most frequent chronic liver disease in Western societies, affecting one in four adults in the USA, and is strongly associated with hepatic insulin resistance, a major risk factor in the pathogenesis of type 2 diabetes. Although the cellular mechanisms underlying this relationship are unknown, hepatic accumulation of diacylglycerol (DAG) in both animals and humans has been linked to hepatic insulin resistance. In this Perspective, we discuss the role of DAG activation of protein kinase Cε as the mechanism responsible for NAFLD-associated hepatic insulin resistance seen in obesity, type 2 diabetes, and lipodystrophy.
Collapse
Affiliation(s)
- François R Jornayvaz
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
| | | |
Collapse
|