1
|
Li QL, Zheng H, Luo Z, Wu LX, Xu PC, Guo JC, Song YF, Tan XY. Characterization and expression analysis of seven lipid metabolism-related genes in yellow catfish Pelteobagrus fulvidraco fed high fat and bile acid diet. Gene 2024; 894:147972. [PMID: 37944648 DOI: 10.1016/j.gene.2023.147972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 09/27/2023] [Accepted: 11/06/2023] [Indexed: 11/12/2023]
Abstract
SREBPs, such as SREBP1 and SREBP2, were the key transcriptional factors regulating lipid metabolism. The processing of SREBPs involved many genes, such as scap, s1p, s2p, cideb. Here, we deciphered the full-length cDNA sequences of scap, srebp1, srebp2, s1p, s2p, cideb and cidec from yellow catfish Pelteobagrus fulvidraco. Their full-length cDNA sequences ranged from 1587 to 3884 bp, and their ORF length from 1191 to 2979 bp, encoding 396-992 amino acids. Some conservative domains were predicted, including the multiple transmembrane domains in SCAP, the bHLH-ZIP domain in SREBP1 and SREBP2, the ApoB binding region, ER targeting region and LD targeting region in CIDEb, the LD targeting region in the CIDEc, the conserved catalytic site and processing site in S1P, and the transmembrane helix domain in S2P. Their mRNA expression could be observed in the heart, spleen, liver, kidney, brain, muscle, intestine and adipose, but varied with tissues. The changes of their mRNA expression in responses to high-fat (HFD) and bile acid (BA) diets were also investigated in the brain, heart, intestine, kidney and spleen tissues. In the brain, HFD significantly increased the mRNA expression of seven genes (scap, srebp1, srebp2, s1p, s2p, cideb and cidec), and the BA attenuated the increase of scap, srebp1, srebp2, s1p, s2p, cideb and cidec mRNA expression induced by HFD. In the heart, HFD significantly increased the mRNA abundances of six genes (srebp1, srebp2, scap, s2p, cideb and cidec), and BA attenuated the increase of their mRNA abundances induced by HFD. In the intestine, HFD increased the cideb, s1p and s2p mRNA abundances, and BA attenuated the HFD-induced increment of their mRNA abundances. In the kidney, HFD significantly increased the scap, cidec and s1p mRNA expression, and BA diet attenuated the increment of their mRNA expression. In the spleen, HFD treatment increased the scap, srebp2, s1p and s2p mRNA expression, and BA diet attenuated HFD-induced increment of their mRNA expression. Taken together, our study elucidated the characterization, expression profiles and transcriptional response of seven lipid metabolic genes, which would serve as the good basis for the further exploration into their function and regulatory mechanism in fish.
Collapse
Affiliation(s)
- Qing-Lin Li
- Laboratory of Molecular Nutrition, Fishery College, Huazhong Agricultural University, Wuhan 430070, China
| | - Hua Zheng
- Laboratory of Molecular Nutrition, Fishery College, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhi Luo
- Laboratory of Molecular Nutrition, Fishery College, Huazhong Agricultural University, Wuhan 430070, China
| | - Li-Xiang Wu
- Laboratory of Molecular Nutrition, Fishery College, Huazhong Agricultural University, Wuhan 430070, China
| | - Peng-Cheng Xu
- Laboratory of Molecular Nutrition, Fishery College, Huazhong Agricultural University, Wuhan 430070, China
| | - Jia-Cheng Guo
- Laboratory of Molecular Nutrition, Fishery College, Huazhong Agricultural University, Wuhan 430070, China
| | - Yu-Feng Song
- Laboratory of Molecular Nutrition, Fishery College, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiao-Ying Tan
- Laboratory of Molecular Nutrition, Fishery College, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
2
|
Zakaria Z, Othman ZA, Nna VU, Mohamed M. The promising roles of medicinal plants and bioactive compounds on hepatic lipid metabolism in the treatment of non-alcoholic fatty liver disease in animal models: molecular targets. Arch Physiol Biochem 2023; 129:1262-1278. [PMID: 34153200 DOI: 10.1080/13813455.2021.1939387] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 06/01/2021] [Indexed: 12/13/2022]
Abstract
Imbalance in hepatic lipid metabolism can lead to an abnormal triglycerides deposition in the hepatocytes which can cause non-alcoholic fatty liver disease (NAFLD). Four main mechanisms responsible for regulating hepatic lipid metabolism are fatty acid uptake, de novo lipogenesis, lipolysis and fatty acid oxidation. Controlling the expression of transcription factors at molecular level plays a crucial role in NAFLD management. This paper reviews various medicinal plants and their bioactive compounds emphasising mechanisms involved in hepatic lipid metabolism, other important NAFLD pathological features, and their promising roles in managing NAFLD through regulating key transcription factors. Although there are many medicinal plants popularly investigated for NAFLD treatment, there is still little information and scientific evidence available and there has been no research on clinical trials scrutinised on this matter. This review also aims to provide molecular information of medicinal plants in NALFD treatment that might have potentials for future scientifically controlled studies.
Collapse
Affiliation(s)
- Zaida Zakaria
- Department of Physiology, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Zaidatul Akmal Othman
- Department of Physiology, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, Kelantan, Malaysia
- Unit of Physiology, Faculty of Medicine, Universiti Sultan Zainal Abidin, Kuala Terengganu, Malaysia
| | - Victor Udo Nna
- Department of Physiology, Faculty of Basic Medical Sciences, College of Medical Sciences, University of Calabar, Calabar, Nigeria
| | - Mahaneem Mohamed
- Department of Physiology, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, Kelantan, Malaysia
- Unit of Integrative Medicine, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, Kelantan, Malaysia
| |
Collapse
|
3
|
Julve J, Genua I, Quifer-Rada P, Yanes Ó, Barranco-Altirriba M, Hernández M, Junza A, Capellades J, Granado-Casas M, Alonso N, Castelblanco E, Mauricio D. Circulating metabolomic and lipidomic changes in subjects with new-onset type 1 diabetes after optimization of glycemic control. Diabetes Res Clin Pract 2023; 197:110578. [PMID: 36804334 DOI: 10.1016/j.diabres.2023.110578] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023]
Abstract
AIMS To uncover novel candidate metabolomic and lipidomic biomarkers in newly-diagnosed type 1 diabetes (T1DM) after achieving optimal glucose control. METHODS Comprehensive lipidomic and metabolomic analysis was performed in serum of 12 adults with T1DM at onset and after achieving optimal glycemic control (HbA1c < 7 %) (after 2-6 months). RESULTS After intensive therapy, subjects (mean age 25.2 years, 58.3 % men) showed decreases in blood glucose (p < 0.001), HbA1c [11.5 % (9.2-13.4) to 6.2 % (5.2 - 6.7); p < 0.001] and changes in 51 identified lipids. Among these changes, we found that triglycerides (TG) containing medium chain fatty acids (TG45:0, TG47:1), sphingomyelins (SM) (SM(d18:2/20:0), SM42:4)), and phosphatidylcholines (PC) (PC(O-26:2), PC(O-30:0), PC(O-32:0), PC(O-42:6), PC(O-44:5), PC(O-38:3), PC(O-33:0), PC(O-46:8), PC(O-44:6), PC(O-40:3), PC(O-42:4), PC(O-46:7), PC(O-46:6), PC(O-44:5), PC(O-42:3), PC(O-44:4)) decreased; whereas PC(35:1), PC(37:1) and TG containing longer chain fatty acids (TG(52:1), TG(55:7), TG(51:2), TG(53:3), TG52:2), TG(53:2), TG(57:3), TG(61:3), TG(61:2) increased. Further, dihydro O-acylceramide (18:1/18:0/16:0), diacylglycerophosphoethanolamine (PE(34:1)), diacylglycerophosphoinositol (PI(38:6), and dihydrosphingomyelins (dihydroSM(36:0), dihydroSM(40:0), dihydroSM(41:0), dihydroSM(42:0)) increased. Uric acid, mannitol, and mannitol-1-acetate levels also increased. CONCLUSIONS Our data uncovered potential favorable changes in the metabolism of glycerophospholipids, glycerolipids, and sphingolipids in new-onset T1DM after achieving optimal glycemic control. Further research on their potential role in developing diabetes-related complications is needed.
Collapse
Affiliation(s)
- Josep Julve
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041 Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Barcelona, Spain
| | - Idoia Genua
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041 Barcelona, Spain; Department of Endocrinology & Nutrition, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Department of Medicine, Autonomous University of Barcelona, Barcelona, Spain
| | - Paola Quifer-Rada
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Barcelona, Spain; LactApp Women's Health, Barcelona, Spain
| | - Óscar Yanes
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Barcelona, Spain; Universitat Rovira i Virgili, Department of Electronic Engineering & IISPV, Tarragona, Spain
| | - Maria Barranco-Altirriba
- Department of Endocrinology & Nutrition, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; B2SLab, Departament d'Enginyeria de Sistemes, Automàtica i Informàtica Industrial, Universitat Politècnica de Catalunya, Barcelona, Spain; Networking Biomedical Research Centre in the Subject Area of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Barcelona, Spain; Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Marta Hernández
- Department of Endocrinology & Nutrition, University Hospital Arnau de Vilanova, Lleida, Spain; Lleida Institute for Biomedical Research Dr. Pifarré Foundation (IRBLleida), University of Lleida, 25198 Lleida, Spain
| | - Alexandra Junza
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Barcelona, Spain; Universitat Rovira i Virgili, Department of Electronic Engineering & IISPV, Tarragona, Spain
| | - Jordi Capellades
- Universitat Rovira i Virgili, Department of Electronic Engineering & IISPV, Tarragona, Spain
| | - Minerva Granado-Casas
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Barcelona, Spain; Lleida Institute for Biomedical Research Dr. Pifarré Foundation (IRBLleida), University of Lleida, 25198 Lleida, Spain
| | - Núria Alonso
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Barcelona, Spain; Department of Medicine, Autonomous University of Barcelona, Barcelona, Spain; Department of Endocrinology & Nutrition, University Hospital Germans Trias i Pujol, 08916 Badalona, Spain
| | - Esmeralda Castelblanco
- Endocrinology, Metabolism and Lipid Research Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Unitat de Suport a la Recerca Barcelona, Institut Universitari d'Investigació en Atenció Primària Jordi Gol i Gurina (IDIAP Jordi Gol), 08007 Barcelona, Spain.
| | - Didac Mauricio
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041 Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Barcelona, Spain; Department of Endocrinology & Nutrition, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Unitat de Suport a la Recerca Barcelona, Institut Universitari d'Investigació en Atenció Primària Jordi Gol i Gurina (IDIAP Jordi Gol), 08007 Barcelona, Spain; Department of Medicine, University of Vic - Central University of Catalonia, Vic, Spain.
| |
Collapse
|
4
|
Iwaki M, Kobayashi T, Nogami A, Saito S, Nakajima A, Yoneda M. Impact of Sarcopenia on Non-Alcoholic Fatty Liver Disease. Nutrients 2023; 15:nu15040891. [PMID: 36839249 PMCID: PMC9965462 DOI: 10.3390/nu15040891] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
With the increasing incidence of non-alcoholic fatty liver disease (NAFLD) and the aging of the population, sarcopenia is attracting attention as one of the pathological conditions involved in the development and progression of NAFLD. In NAFLD, sarcopenia is closely associated with insulin resistance and results from the atrophy of skeletal muscle, an insulin target organ. In addition, inflammatory cytokines that promote skeletal muscle protein breakdown, low adiponectin levels leading to decreased insulin sensitivity, and hyperleptinemia are also involved in NAFLD pathogenesis. The presence of sarcopenia is a prognostic factor and increases the risk of mortality in patients with cirrhosis and post-treatment liver cancer. Sarcopenia, the presence of which mainly occurs due to decreased muscle mass, combined with increased visceral fat, can lead to sarcopenia-associated obesity, which increases the risk of NASH, liver fibrosis, and cardiovascular disease. In order to treat sarcopenia, it is necessary to properly evaluate sarcopenia status. Patients with high BMI, as in sarcopenic obesity, may improve with caloric restriction. However, inadequate oral intake may lead to further loss of muscle mass. Aerobic and resistance exercise should also be used appropriately.
Collapse
|
5
|
Long-term statins administration exacerbates diabetic nephropathy via ectopic fat deposition in diabetic mice. Nat Commun 2023; 14:390. [PMID: 36693830 PMCID: PMC9873739 DOI: 10.1038/s41467-023-35944-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/09/2023] [Indexed: 01/25/2023] Open
Abstract
Statins play an important role in the treatment of diabetic nephropathy. Increasing attention has been given to the relationship between statins and insulin resistance, but many randomized controlled trials confirm that the therapeutic effects of statins on diabetic nephropathy are more beneficial than harmful. However, further confirmation of whether the beneficial effects of chronic statin administration on diabetic nephropathy outweigh the detrimental effects is urgently needed. Here, we find that long-term statin administration may increase insulin resistance, interfere with lipid metabolism, leads to inflammation and fibrosis, and ultimately fuel diabetic nephropathy progression in diabetic mice. Mechanistically, activation of insulin-regulated phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin signaling pathway leads to increased fatty acid synthesis. Furthermore, statins administration increases lipid uptake and inhibits fatty acid oxidation, leading to lipid deposition. Here we show that long-term statins administration exacerbates diabetic nephropathy via ectopic fat deposition in diabetic mice.
Collapse
|
6
|
Dong Y, Liu L, Li M, Xie D, Zhao J, Wang S, You C, Li Y. Insulin can up-regulate LC-PUFA biosynthesis with the involvement of Srebp-1c and stimulatory protein 1 (Sp1) in marine teleost Siganus canaliculatus. Gene X 2022; 840:146755. [PMID: 35905852 DOI: 10.1016/j.gene.2022.146755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/13/2022] [Accepted: 07/24/2022] [Indexed: 11/04/2022] Open
Abstract
The rabbitfish Siganus canaliculatus is the first marine teleost found to have the biosynthetic ability of long-chain polyunsaturated fatty acids (LC-PUFA) from C18 precursors catalyzed by fatty acyl desaturases (Δ6/Δ5 Fads, Δ4 Fads) and elongases of very long chain fatty acids (Elovls). Previously, we predicted the existence of insulin (INS) response elements (IREs) including nuclear factor Y (NF-Y) and sterol regulatory element (SRE) in the core promoter region of rabbitfish Δ6/Δ5 fads and Δ4 fads. To clarify the potential regulatory effect and mechanism of INS in LC-PUFA biosynthesis, INS responding region was identified at -456 bp to + 51 bp of Δ6/Δ5 fads core promoter, but not in Δ4 fads promoter. Moreover, a unique stimulatory protein 1 (Sp1) element was predicted in the INS responding region of Δ6/Δ5 fads. Subsequently, SRE, NF-Y and Sp1 elements were proved as IREs in Δ6/Δ5 fads promoter. The up-regulation of INS on gene expression of Srebp-1c, Sp1, Δ6/Δ5 fads and elovl5 as well as the LC-PUFA biosynthesis was further demonstrated in S. canaliculatus hepatocyte line (SCHL) cells, but no influence was detected on Δ4 fads. Besides, inhibitors of transcription factors Srebp-1c (Fatostatin, PF-429242) and Sp1 (Mithramycin) could inhibit the gene expression of Srebp-1c, Δ6/Δ5 fads and elovl5, and abolish the up-regulation of INS on these genes' expression and LC-PUFA biosynthesis. These results indicated that INS could up-regulate LC-PUFA biosynthesis with the involvement of Srebp-1c and Sp1 in rabbitfish S. canaliculatus, which is the first report in teleost.
Collapse
Affiliation(s)
- Yewei Dong
- College of Marine Sciences of South China Agricultural University & Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China; College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, 510225, China
| | - Lijie Liu
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, Guangdong, 515063, China
| | - Mengmeng Li
- College of Marine Sciences of South China Agricultural University & Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Dizhi Xie
- College of Marine Sciences of South China Agricultural University & Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Jianhong Zhao
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, Guangdong, 515063, China
| | - Shuqi Wang
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, Guangdong, 515063, China
| | - Cuihong You
- College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, 510225, China
| | - Yuanyou Li
- College of Marine Sciences of South China Agricultural University & Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China.
| |
Collapse
|
7
|
Videla LA, Hernandez-Rodas MC, Metherel AH, Valenzuela R. Influence of the nutritional status and oxidative stress in the desaturation and elongation of n-3 and n-6 polyunsaturated fatty acids: Impact on non-alcoholic fatty liver disease. Prostaglandins Leukot Essent Fatty Acids 2022; 181:102441. [PMID: 35537354 DOI: 10.1016/j.plefa.2022.102441] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 12/25/2022]
Abstract
Polyunsaturated fatty acids (PUFA) play essential roles in cell membrane structure and physiological processes including signal transduction, cellular metabolism and tissue homeostasis to combat diseases. PUFA are either consumed from food or synthesized by enzymatic desaturation, elongation and peroxisomal β-oxidation. The nutritionally essential precursors α-linolenic acid (C18:3n-3; ALA) and linoleic acid (C18:2n-6; LA) are subjected to desaturation by Δ6D/Δ5D desaturases and elongation by elongases 2/5, enzymes that are induced by insulin and repressed by PUFA. Maintaining an optimally low n-6/n-3 PUFA ratio is linked to prevention of the development of several diseases, including nonalcoholic fatty liver disease (NAFLD) that is characterized by depletion of PUFA promoting hepatic steatosis and inflammation. In this context, supplementation with n-3 PUFA revealed significant lowering of hepatic steatosis in obese patients, whereas prevention of fatty liver by high-fat diet in mice is observed in n-3 PUFA and hydroxytyrosol co-administration. The aim of this work is to review the role of nutritional status and nutrient availability on markers of PUFA biosynthesis. In addition, the impact of oxidative stress developed as a result of NAFLD, a redox imbalance that may alter the expression and activity of the enzymes involved, and diminished n-3 PUFA levels by free-radical dependent peroxidation processes will be discussed.
Collapse
Affiliation(s)
- Luis A Videla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | | | - Adam H Metherel
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Rodrigo Valenzuela
- Nutrition Department, Faculty of Medicine, University of Chile, Santiago, Chile; Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
8
|
Adimulam T, Abdul N, Chuturgoon A. HepG2 liver cells treated with fumonisin B1 in galactose supplemented media have altered expression of genes and proteins known to regulate cholesterol flux. WORLD MYCOTOXIN J 2021. [DOI: 10.3920/wmj2021.2723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Fumonisin B1 (FB1) contributes to mycotoxicosis in animals and has been associated with the incidence of some cancers in humans. The effect of FB1 on lipidomic profiles, sphingolipids and cholesterol levels have been demonstrated in experimental models, however, the events leading to altered cholesterol levels are unclear. This study investigates the molecular mechanisms that regulate the effect of FB1 on cholesterol homeostasis in galactose supplemented HepG2 liver cells. Galactose supplementation is a proven method utilised to circumvent the Crabtree effect exhibited by cancer cells, which forces cancer cells to activate the mitochondria. HepG2 cells were cultured in galactose supplemented media and treated with FB1 (IC50 = 25 μM) for 6 h. Cell viability was determined using the MTT assay. Metabolic status was evaluated using ATP luciferase assay, and cholesterol regulatory transcription factors (SIRT1, SREBP-1C, LXR, LDLR, PCSK9, and ABCA1) were investigated using western blotting and qPCR. FB1 in galactose supplemented HepG2 cells increased gene expression of SIRT1 (P<0.05), SREBP-1C, LXR, and LDLR; however, PCSK9 (P<0.05) was decreased. Furthermore, protein expression of SIRT1, LXR, and LDLR was elevated upon FB1 treatment, while SREBP-1C and PCSK9 were reduced. The data provides evidence that SIRT1 reduced the expression of PCSK9 and deacetylated LXR to prevent degradation of LDLR. This could result in a dysregulated cholesterol flux, which may contribute to FB1 mediated toxicity.
Collapse
Affiliation(s)
- T. Adimulam
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4041, South Africa
| | - N.S. Abdul
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4041, South Africa
- Applied Microbial and Health Biotechnology Institute, Cape Peninsula University of Technology, Cape Town 7535, South Africa
| | - A.A. Chuturgoon
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4041, South Africa
| |
Collapse
|
9
|
Insulin-Responsive Transcription Factors. Biomolecules 2021; 11:biom11121886. [PMID: 34944530 PMCID: PMC8699568 DOI: 10.3390/biom11121886] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/04/2021] [Accepted: 12/09/2021] [Indexed: 12/12/2022] Open
Abstract
The hormone insulin executes its function via binding and activating of the insulin receptor, a receptor tyrosine kinase that is mainly expressed in skeletal muscle, adipocytes, liver, pancreatic β-cells, and in some areas of the central nervous system. Stimulation of the insulin receptor activates intracellular signaling cascades involving the enzymes extracellular signal-regulated protein kinase-1/2 (ERK1/2), phosphatidylinositol 3-kinase, protein kinase B/Akt, and phospholipase Cγ as signal transducers. Insulin receptor stimulation is correlated with multiple physiological and biochemical functions, including glucose transport, glucose homeostasis, food intake, proliferation, glycolysis, and lipogenesis. This review article focuses on the activation of gene transcription as a result of insulin receptor stimulation. Signal transducers such as protein kinases or the GLUT4-induced influx of glucose connect insulin receptor stimulation with transcription. We discuss insulin-responsive transcription factors that respond to insulin receptor activation and generate a transcriptional network executing the metabolic functions of insulin. Importantly, insulin receptor stimulation induces transcription of genes encoding essential enzymes of glycolysis and lipogenesis and inhibits genes encoding essential enzymes of gluconeogenesis. Overall, the activation or inhibition of insulin-responsive transcription factors is an essential aspect of orchestrating a wide range of insulin-induced changes in the biochemistry and physiology of insulin-responsive tissues.
Collapse
|
10
|
Yu S, Meng S, Xiang M, Ma H. Phosphoenolpyruvate carboxykinase in cell metabolism: Roles and mechanisms beyond gluconeogenesis. Mol Metab 2021; 53:101257. [PMID: 34020084 PMCID: PMC8190478 DOI: 10.1016/j.molmet.2021.101257] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Phosphoenolpyruvate carboxykinase (PCK) has been almost exclusively recognized as a critical enzyme in gluconeogenesis, especially in the liver and kidney. Accumulating evidence has shown that the enhanced activity of PCK leads to increased glucose output and exacerbation of diabetes, whereas the defects of PCK result in lethal hypoglycemia. Genetic mutations or polymorphisms are reported to be related to the onset and progression of diabetes in humans. SCOPE OF REVIEW Recent studies revealed that the PCK pathway is more complex than just gluconeogenesis, depending on the health or disease condition. Dysregulation of PCK may contribute to the development of obesity, cardiac hypertrophy, stroke, and cancer. Moreover, a regulatory network with multiple layers, from epigenetic regulation, transcription regulation, to posttranscription regulation, precisely tunes the expression of PCK. Deciphering the molecular basis that regulates PCK may pave the way for developing practical strategies to treat metabolic dysfunction. MAJOR CONCLUSIONS In this review, we summarize the metabolic and non-metabolic roles of the PCK enzyme in cells, especially beyond gluconeogenesis. We highlight the distinct functions of PCK isoforms (PCK1 and PCK2), depict a detailed network regulating PCK's expression, and discuss its clinical relevance. We also discuss the therapeutic potential targeting PCK and the future direction that is highly in need to better understand PCK-mediated signaling under diverse conditions.
Collapse
Affiliation(s)
- Shuo Yu
- Anesthesiology Department, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Simin Meng
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Meixiang Xiang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China.
| | - Hong Ma
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China.
| |
Collapse
|
11
|
SREBP-1c and lipogenesis in the liver: an update1. Biochem J 2021; 478:3723-3739. [PMID: 34673919 DOI: 10.1042/bcj20210071] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/28/2021] [Accepted: 09/30/2021] [Indexed: 12/13/2022]
Abstract
Sterol Regulatory Element Binding Protein-1c is a transcription factor that controls the synthesis of lipids from glucose in the liver, a process which is of utmost importance for the storage of energy. Discovered in the early nineties by B. Spiegelman and by M. Brown and J. Goldstein, it has generated more than 5000 studies in order to elucidate its mechanism of activation and its role in physiology and pathology. Synthetized as a precursor found in the membranes of the endoplasmic reticulum, it has to be exported to the Golgi and cleaved by a mechanism called regulated intramembrane proteolysis. We reviewed in 2002 its main characteristics, its activation process and its role in the regulation of hepatic glycolytic and lipogenic genes. We particularly emphasized that Sterol Regulatory Element Binding Protein-1c is the mediator of insulin effects on these genes. In the present review, we would like to update these informations and focus on the response to insulin and to another actor in Sterol Regulatory Element Binding Protein-1c activation, the endoplasmic reticulum stress.
Collapse
|
12
|
Lee K, Yu H, Shouse S, Kong B, Lee J, Lee SH, Ko KS. RNA-Seq Reveals Different Gene Expression in Liver-Specific Prohibitin 1 Knock-Out Mice. Front Physiol 2021; 12:717911. [PMID: 34539442 PMCID: PMC8446661 DOI: 10.3389/fphys.2021.717911] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/27/2021] [Indexed: 12/24/2022] Open
Abstract
Prohibitin 1 (PHB1) is an evolutionarily conserved and ubiquitously expressed protein that stabilizes mitochondrial chaperone. Our previous studies showed that liver-specific Phb1 deficiency induced liver injuries and aggravated lipopolysaccharide (LPS)-induced innate immune responses. In this study, we performed RNA-sequencing (RNA-seq) analysis with liver tissues to investigate global gene expression among liver-specific Phb1−/−, Phb1+/−, and WT mice, focusing on the differentially expressed (DE) genes between Phb1+/− and WT. When 78 DE genes were analyzed for biological functions, using ingenuity pathway analysis (IPA) tool, lipid metabolism-related genes, including insulin receptor (Insr), sterol regulatory element-binding transcription factor 1 (Srebf1), Srebf2, and SREBP cleavage-activating protein (Scap) appeared to be downregulated in liver-specific Phb1+/− compared with WT. Diseases and biofunctions analyses conducted by IPA verified that hepatic system diseases, including liver fibrosis, liver hyperplasia/hyperproliferation, and liver necrosis/cell death, which may be caused by hepatotoxicity, were highly associated with liver-specific Phb1 deficiency in mice. Interestingly, of liver disease-related 5 DE genes between Phb1+/− and WT, the mRNA expressions of forkhead box M1 (Foxm1) and TIMP inhibitor of metalloproteinase (Timp1) were matched with validation for RNA-seq in liver tissues and AML12 cells transfected with Phb1 siRNA. The results in this study provide additional insights into molecular mechanisms responsible for increasing susceptibility of liver injuries associated with hepatic Phb1.
Collapse
Affiliation(s)
- Kyuwon Lee
- Department of Nutritional Science and Food Management, College of Science and Industry Convergence, Ewha Womans University, Seoul, South Korea
| | - Hyeonju Yu
- Department of Nutritional Science and Food Management, College of Science and Industry Convergence, Ewha Womans University, Seoul, South Korea
| | - Stephanie Shouse
- Center of Excellence for Poultry Science, University of Arkansas System Division of Agriculture, Fayetteville, AR, United States
| | - Byungwhi Kong
- Center of Excellence for Poultry Science, University of Arkansas System Division of Agriculture, Fayetteville, AR, United States
| | - Jihye Lee
- Department of Nutrition and Food Science, College of Agriculture and Natural Resources, University of Maryland, College Park, MD, United States
| | - Seong-Ho Lee
- Department of Nutrition and Food Science, College of Agriculture and Natural Resources, University of Maryland, College Park, MD, United States
| | - Kwang Suk Ko
- Department of Nutritional Science and Food Management, College of Science and Industry Convergence, Ewha Womans University, Seoul, South Korea.,Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Beverly Hills, CA, United States
| |
Collapse
|
13
|
Moldavski O, Zushin PJH, Berdan CA, Van Eijkeren RJ, Jiang X, Qian M, Ory DS, Covey DF, Nomura DK, Stahl A, Weiss EJ, Zoncu R. 4β-Hydroxycholesterol is a prolipogenic factor that promotes SREBP1c expression and activity through the liver X receptor. J Lipid Res 2021; 62:100051. [PMID: 33631213 PMCID: PMC8042401 DOI: 10.1016/j.jlr.2021.100051] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 01/06/2021] [Accepted: 02/12/2021] [Indexed: 12/16/2022] Open
Abstract
Oxysterols are oxidized derivatives of cholesterol that play regulatory roles in lipid biosynthesis and homeostasis. How oxysterol signaling coordinates different lipid classes such as sterols and triglycerides remains incompletely understood. Here, we show that 4β-hydroxycholesterol (HC) (4β-HC), a liver and serum abundant oxysterol of poorly defined functions, is a potent and selective inducer of the master lipogenic transcription factor, SREBP1c, but not the related steroidogenic transcription factor SREBP2. By correlating tracing of lipid synthesis with lipogenic gene expression profiling, we found that 4β-HC acts as a putative agonist for the liver X receptor (LXR), a sterol sensor and transcriptional regulator previously linked to SREBP1c activation. Unique among the oxysterol agonists of the LXR, 4β-HC induced expression of the lipogenic program downstream of SREBP1c and triggered de novo lipogenesis both in primary hepatocytes and in the mouse liver. In addition, 4β-HC acted in parallel to insulin-PI3K-dependent signaling to stimulate triglyceride synthesis and lipid-droplet accumulation. Thus, 4β-HC is an endogenous regulator of de novo lipogenesis through the LXR-SREBP1c axis.
Collapse
Affiliation(s)
- Ofer Moldavski
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA, USA; The Paul F. Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA, USA; Cardiovascular Research Institute, UCSF, San Francisco, CA, USA
| | - Peter-James H Zushin
- Department of Nutritional Sciences and Toxicology, University of California at Berkeley, Berkeley, CA, USA
| | - Charles A Berdan
- Department of Nutritional Sciences and Toxicology, University of California at Berkeley, Berkeley, CA, USA
| | - Robert J Van Eijkeren
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA, USA; The Paul F. Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA, USA
| | - Xuntian Jiang
- Diabetic Cardiovascular Disease Center, Washington University School of Medicine, St Louis, MO, USA
| | - Mingxing Qian
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Daniel S Ory
- Diabetic Cardiovascular Disease Center, Washington University School of Medicine, St Louis, MO, USA
| | - Douglas F Covey
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Daniel K Nomura
- Department of Nutritional Sciences and Toxicology, University of California at Berkeley, Berkeley, CA, USA
| | - Andreas Stahl
- Department of Nutritional Sciences and Toxicology, University of California at Berkeley, Berkeley, CA, USA
| | - Ethan J Weiss
- Cardiovascular Research Institute, UCSF, San Francisco, CA, USA
| | - Roberto Zoncu
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA, USA; The Paul F. Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA, USA.
| |
Collapse
|
14
|
Mertens J, Van Gaal LF, Francque SM, De Block C. NAFLD in type 1 diabetes: overrated or underappreciated? Ther Adv Endocrinol Metab 2021; 12:20420188211055557. [PMID: 34840719 PMCID: PMC8613893 DOI: 10.1177/20420188211055557] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 10/04/2021] [Indexed: 12/18/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease in western countries, affecting 25-30% of the general population and up to 65% in those with obesity and/or type 2 diabetes. Accumulation of visceral adipose tissue and insulin resistance (IR) contributes to NAFLD. NAFLD is not an innocent entity as it not only may cause nonalcoholic steatohepatitis and cirrhosis but also contribute to cardiovascular morbidity and mortality. More and more people with type 1 diabetes (T1D) are becoming overweight and present with features of IR, but the prevalence and impact of NAFLD in this population are still unclear. The utility of noninvasive screening tools for NAFLD in T1D is being explored. Recent data indicate that based upon ultrasonographic criteria NAFLD is present in 27% (ranging between 19% and 31%) of adults with T1D. Magnetic resonance imaging data indicate a prevalence rate of 8.6% (ranging between 2.1% and 18.6%). There are, however, multiple factors affecting these data, ranging from study design and referral bias to discrepancies in between diagnostic modalities. Individuals with T1D have a 7-fold higher risk of cardiovascular disease (CVD) and cardiovascular mortality is the most prominent cause of death in T1D. Patients with T1D and NALFD are also more prone to develop CVD, but the independent contribution of NAFLD to cardiovascular events has to be determined in this population. Furthermore, limited data in T1D also point towards a 2 to 3 times higher risk for microvascular complications in those with NAFLD. In this article, we will discuss epidemiological and diagnostic challenges of NAFLD in T1D, explore the link between IR and NAFLD and chronic complications, and examine the independent contribution of NAFLD to the presence of macro-, and microvascular complications.
Collapse
Affiliation(s)
- Jonathan Mertens
- Department of Endocrinology, Diabetology and Metabolism, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium
- Laboratory of Experimental Medicine and Pediatrics and Member of the Infla-Med Centre of Excellence, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
- Department of Gastroenterology & Hepatology, Antwerp University Hospital, Edegem, Belgium
| | - Luc F. Van Gaal
- Department of Endocrinology, Diabetology and Metabolism, Antwerp University Hospital, Edegem, Belgium
- Laboratory of Experimental Medicine and Pediatrics and Member of the Infla-Med Centre of Excellence, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Sven M. Francque
- Department of Gastroenterology & Hepatology, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium
- Laboratory of Experimental Medicine and Pediatrics and Member of the Infla-Med Centre of Excellence, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | | |
Collapse
|
15
|
Manka PP, Kaya E, Canbay A, Syn WK. A Review of the Epidemiology, Pathophysiology, and Efficacy of Anti-diabetic Drugs Used in the Treatment of Nonalcoholic Fatty Liver Disease. Dig Dis Sci 2021; 66:3676-3688. [PMID: 34410573 PMCID: PMC8510897 DOI: 10.1007/s10620-021-07206-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/22/2021] [Indexed: 02/06/2023]
Abstract
In recent years, epidemiological studies have consistently demonstrated that the coexistence of nonalcoholic fatty liver disease (NAFLD) and type 2 diabetes mellitus (T2DM) is strongly associated with increased mortality and morbidity related to hepatic- and extrahepatic causes. Indeed, compared with the general population, patients with T2DM are more likely to be diagnosed with more severe forms of NAFLD (i.e., nonalcoholic steatohepatitis (NASH) with liver fibrosis). There is an ongoing debate whether NALFD is a consequence of diabetes or whether NAFLD is simply a component and manifestation of the metabolic syndrome, since liver fat (steatosis) and even more advanced stages of liver fibrosis can occur in the absence of diabetes. Nevertheless, insulin resistance is a key component of the mechanism of NAFLD development; furthermore, therapies that lower blood glucose concentrations also appear to be effective in the treatment of NAFLD. Here, we will discuss the pathophysiological and epidemiological associations between NAFLD and T2DM. We will also review currently available anti-diabetic agents with their regard to their efficacy of NAFLD/NASH treatment.
Collapse
Affiliation(s)
- Paul P. Manka
- grid.5570.70000 0004 0490 981XDepartment of Internal Medicine, University Hospital Knappschaftskrankenhaus, Ruhr-University Bochum, In der Schornau 23-25, 44892 Bochum, Germany
| | - Eda Kaya
- grid.5570.70000 0004 0490 981XDepartment of Internal Medicine, University Hospital Knappschaftskrankenhaus, Ruhr-University Bochum, In der Schornau 23-25, 44892 Bochum, Germany
| | - Ali Canbay
- grid.5570.70000 0004 0490 981XDepartment of Internal Medicine, University Hospital Knappschaftskrankenhaus, Ruhr-University Bochum, In der Schornau 23-25, 44892 Bochum, Germany
| | - Wing-Kin Syn
- grid.259828.c0000 0001 2189 3475Division of Gastroenterology and Hepatology, Medical University of South Carolina, Charleston, SC USA ,grid.11480.3c0000000121671098Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country, Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), Leioa, Spain ,grid.280644.c0000 0000 8950 3536Section of Gastroenterology, Ralph H Johnson Veterans Affairs Medical Center, Charleston, SC USA
| |
Collapse
|
16
|
Allen JN, Dey A, Cai J, Zhang J, Tian Y, Kennett M, Ma Y, Liang TJ, Patterson AD, Hankey-Giblin PA. Metabolic Profiling Reveals Aggravated Non-Alcoholic Steatohepatitis in High-Fat High-Cholesterol Diet-Fed Apolipoprotein E-Deficient Mice Lacking Ron Receptor Signaling. Metabolites 2020; 10:metabo10080326. [PMID: 32796650 PMCID: PMC7464030 DOI: 10.3390/metabo10080326] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/29/2020] [Accepted: 08/03/2020] [Indexed: 12/13/2022] Open
Abstract
Non-alcoholic steatohepatitis (NASH) represents the progressive sub-disease of non-alcoholic fatty liver disease that causes chronic liver injury initiated and sustained by steatosis and necroinflammation. The Ron receptor is a tyrosine kinase of the Met proto-oncogene family that potentially has a beneficial role in adipose and liver-specific inflammatory responses, as well as glucose and lipid metabolism. Since its discovery two decades ago, the Ron receptor has been extensively investigated for its differential roles on inflammation and cancer. Previously, we showed that Ron expression on tissue-resident macrophages limits inflammatory macrophage activation and promotes a repair phenotype, which can retard the progression of NASH in a diet-induced mouse model. However, the metabolic consequences of Ron activation have not previously been investigated. Here, we explored the effects of Ron receptor activation on major metabolic pathways that underlie the development and progression of NASH. Mice lacking apolipoprotein E (ApoE KO) and double knockout (DKO) mice that lack ApoE and Ron were maintained on a high-fat high-cholesterol diet for 18 weeks. We observed that, in DKO mice, the loss of ligand-dependent Ron signaling aggravated key pathological features in steatohepatitis, including steatosis, inflammation, oxidation stress, and hepatocyte damage. Transcriptional programs positively regulating fatty acid (FA) synthesis and uptake were upregulated in the absence of Ron receptor signaling, whereas lipid disposal pathways were downregulated. Consistent with the deregulation of lipid metabolism pathways, the DKO animals exhibited increased accumulation of FAs in the liver and decreased level of bile acids. Altogether, ligand-dependent Ron receptor activation provides protection from the deregulation of major metabolic pathways that initiate and aggravate non-alcoholic steatohepatitis.
Collapse
Affiliation(s)
- Joselyn N. Allen
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.N.A.); (A.D.); (J.C.); (J.Z.); (Y.T.); (M.K.)
| | - Adwitia Dey
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.N.A.); (A.D.); (J.C.); (J.Z.); (Y.T.); (M.K.)
| | - Jingwei Cai
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.N.A.); (A.D.); (J.C.); (J.Z.); (Y.T.); (M.K.)
| | - Jingtao Zhang
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.N.A.); (A.D.); (J.C.); (J.Z.); (Y.T.); (M.K.)
| | - Yuan Tian
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.N.A.); (A.D.); (J.C.); (J.Z.); (Y.T.); (M.K.)
| | - Mary Kennett
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.N.A.); (A.D.); (J.C.); (J.Z.); (Y.T.); (M.K.)
| | - Yanling Ma
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, The National Institutes of Health, Bethesda, MD 20814, USA; (Y.M.); (T.J.L.)
| | - T. Jake Liang
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, The National Institutes of Health, Bethesda, MD 20814, USA; (Y.M.); (T.J.L.)
| | - Andrew D. Patterson
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.N.A.); (A.D.); (J.C.); (J.Z.); (Y.T.); (M.K.)
- Correspondence: (A.D.P.); (P.A.H.-G.); Tel.: +1-814-867-4565; (A.D.P.); +1-814-863-0128 (P.A.H.-G.)
| | - Pamela A. Hankey-Giblin
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.N.A.); (A.D.); (J.C.); (J.Z.); (Y.T.); (M.K.)
- Correspondence: (A.D.P.); (P.A.H.-G.); Tel.: +1-814-867-4565; (A.D.P.); +1-814-863-0128 (P.A.H.-G.)
| |
Collapse
|
17
|
Jiang X, Zheng J, Zhang S, Wang B, Wu C, Guo X. Advances in the Involvement of Gut Microbiota in Pathophysiology of NAFLD. Front Med (Lausanne) 2020; 7:361. [PMID: 32850884 PMCID: PMC7403443 DOI: 10.3389/fmed.2020.00361] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 06/15/2020] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is characterized by hepatic steatosis and progresses to non-steatohepatitis (NASH) when the liver displays overt inflammatory damage. Increasing evidence has implicated critical roles for dysbiosis and microbiota-host interactions in NAFLD pathophysiology. In particular, microbiota alter intestine absorption of nutrients and intestine permeability, whose dysregulation enhances the delivery of nutrients, endotoxin, and microbiota metabolites to the liver and exacerbates hepatic fat deposition and inflammation. While how altered composition of gut microbiota attributes to NAFLD remains to be elucidated, microbiota metabolites are shown to be involved in the regulation of hepatocyte fat metabolism and liver inflammatory responses. In addition, intestinal microbes and circadian coordinately adjust metabolic regulation in different stages of life. During aging, altered composition of gut microbiota, along with circadian clock dysregulation, appears to contribute to increased incidence and/or severity of NAFLD.
Collapse
Affiliation(s)
- Xiaofan Jiang
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Juan Zheng
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| | - Shixiu Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Baozhen Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chaodong Wu
- Department of Nutrition, Texas A&M University, College Station, TX, United States
| | - Xin Guo
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
18
|
Seenappa V, Joshi MB, Satyamoorthy K. Intricate Regulation of Phosphoenolpyruvate Carboxykinase (PEPCK) Isoforms in Normal Physiology and Disease. Curr Mol Med 2020; 19:247-272. [PMID: 30947672 DOI: 10.2174/1566524019666190404155801] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 03/25/2019] [Accepted: 03/27/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND The phosphoenolpyruvate carboxykinase (PEPCK) isoforms are considered as rate-limiting enzymes for gluconeogenesis and glyceroneogenesis pathways. PEPCK exhibits several interesting features such as a) organelle-specific isoforms (cytosolic and a mitochondrial) in vertebrate clade, b) tissue-specific expression of isoforms and c) organism-specific requirement of ATP or GTP as a cofactor. In higher organisms, PEPCK isoforms are intricately regulated and activated through several physiological and pathological stimuli such as corticoids, hormones, nutrient starvation and hypoxia. Isoform-specific transcriptional/translational regulation and their interplay in maintaining glucose homeostasis remain to be fully understood. Mounting evidence indicates the significant involvement of PEPCK isoforms in physiological processes (development and longevity) and in the progression of a variety of diseases (metabolic disorders, cancer, Smith-Magenis syndrome). OBJECTIVE The present systematic review aimed to assimilate existing knowledge of transcriptional and translational regulation of PEPCK isoforms derived from cell, animal and clinical models. CONCLUSION Based on current knowledge and extensive bioinformatics analysis, in this review we have provided a comparative (epi)genetic understanding of PCK1 and PCK2 genes encompassing regulatory elements, disease-associated polymorphisms, copy number variations, regulatory miRNAs and CpG densities. We have also discussed various exogenous and endogenous modulators of PEPCK isoforms and their signaling mechanisms. A comprehensive review of existing knowledge of PEPCK regulation and function may enable identification of the underlying gaps to design new pharmacological strategies and interventions for the diseases associated with gluconeogenesis.
Collapse
Affiliation(s)
- Venu Seenappa
- School of Life Sciences, Manipal Academy of Higher Education, Manipal - 576104, India
| | - Manjunath B Joshi
- School of Life Sciences, Manipal Academy of Higher Education, Manipal - 576104, India
| | - Kapaettu Satyamoorthy
- School of Life Sciences, Manipal Academy of Higher Education, Manipal - 576104, India
| |
Collapse
|
19
|
Giudetti AM, Micioni Di Bonaventura MV, Ferramosca A, Longo S, Micioni Di Bonaventura E, Friuli M, Romano A, Gaetani S, Cifani C. Brief daily access to cafeteria-style diet impairs hepatic metabolism even in the absence of excessive body weight gain in rats. FASEB J 2020; 34:9358-9371. [PMID: 32463138 DOI: 10.1096/fj.201902757r] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 04/22/2020] [Accepted: 04/30/2020] [Indexed: 02/06/2023]
Abstract
Numerous nutritional approaches aimed at reducing body weight have been developed as a strategy to reduce obesity. Most of these interventions rely on reducing caloric intake or limiting calories access to a few hours per day. In this work, we analyzed the effects of the extended (24 hours/day) or restricted (1 hour/day) access to a cafeteria-style (CAF) diet, on rat body weight and hepatic lipid metabolism, with respect to control rats (CTR) fed with a standard chow diet. The body weight gain of restricted-fed rats was not different from CTR, despite the slightly higher total caloric intake, but resulted significantly lower than extended-fed rats, which showed a CAF diet-induced obesity and a dramatically higher total caloric intake. However, both CAF-fed groups of rats showed, compared to CTR, unhealthy serum and hepatic parameters such as higher serum glucose level, lower HDL values, and increased hepatic triacylglycerol and cholesterol amount. The hepatic expression and activity of key enzymes of fatty acid synthesis, acetyl-CoA carboxylase (ACC), and fatty acid synthase (FAS), was similarly reduced in both CAF-fed groups of rats with respect to CTR. Anyway, while in extended-fed rats this reduction was associated to a long-term mechanism involving sterol regulatory element-binding protein-1 (SREBP-1), in restricted-fed animals a short-term mechanism based on PKA and AMPK activation occurred in the liver. Furthermore, hepatic fatty acid oxidation (FAO) and oxidative stress resulted significantly increased in extended, but not in restricted-fed rats, as compared to CTR. Overall, these results demonstrate that although limiting the total caloric intake might successfully fight obesity development, the nutritional content of the diet is the major determinant for the health status.
Collapse
Affiliation(s)
- Anna Maria Giudetti
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | | | - Alessandra Ferramosca
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Serena Longo
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | | | - Marzia Friuli
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| | - Adele Romano
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| | - Silvana Gaetani
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| | - Carlo Cifani
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| |
Collapse
|
20
|
Lee H, Kong G, Tran Q, Kim C, Park J, Park J. Relationship Between Ginsenoside Rg3 and Metabolic Syndrome. Front Pharmacol 2020; 11:130. [PMID: 32161549 PMCID: PMC7052819 DOI: 10.3389/fphar.2020.00130] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 01/30/2020] [Indexed: 02/06/2023] Open
Abstract
Metabolic syndrome is an important public health issue and is associated with a more affluent lifestyle. Many studies of metabolic syndrome have been reported, but its pathogenesis remains unclear and there is no effective treatment. The ability of natural compounds to ameliorate metabolic syndrome is currently under investigation. Unlike synthetic chemicals, such natural products have proven utility in various fields. Recently, ginsenoside extracted from ginseng and ginseng root are representative examples. For example, ginseng is used in dietary supplements and cosmetics. In addition, various studies have reported the effects of ginsenoside on metabolic syndromes such as obesity, diabetes, and hypertension. In this review, we describe the potential of ginsenoside Rg3, a component of ginseng, in the treatment of metabolic syndrome.
Collapse
Affiliation(s)
- Hyunji Lee
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, South Korea.,Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Gyeyeong Kong
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, South Korea.,Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Quangdon Tran
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, South Korea.,Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Chaeyeong Kim
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, South Korea.,Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Jisoo Park
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, South Korea.,Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, South Korea.,Department of Life Science, Hyehwa Liberal Arts College, Daejeon University, Daejeon, South Korea
| | - Jongsun Park
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, South Korea.,Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, South Korea
| |
Collapse
|
21
|
Vergani L. Fatty Acids and Effects on In Vitro and In Vivo Models of Liver Steatosis. Curr Med Chem 2019; 26:3439-3456. [PMID: 28521680 DOI: 10.2174/0929867324666170518101334] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 03/14/2017] [Accepted: 03/14/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND Fatty liver, or steatosis, is a condition of excess accumulation of lipids, mainly under form of triglycerides (TG), in the liver, and it is the hallmark of non-alcoholic fatty liver disease (NAFLD). NAFLD is the most common liver disorder world-wide and it has frequently been associated with obesity, hyperlipidemia and insulin resistance. Free fatty acids (FA) are the major mediators of hepatic steatosis; patients with NAFLD have elevated levels of circulating FA that correlate with disease severity. METHODS Steatosis is a reversible condition that can be resolved with changed behaviors, or that can progress towards more severe liver damages such as steatohepatitis (NASH), fibrosis and cirrhosis. In NAFLD, FA of exogenous or endogenous origin accumulate in the hepatocytes and trigger liver damages. Excess TG are stored in cytosolic lipid droplets (LDs) that are dynamic organelles acting as hubs for lipid metabolism. RESULTS In the first part of this review, we briefly reassumed the main classes of FA and their chemical classification as a function of the presence and number of double bonds, their metabolic pathways and effects on human health. Then, we summarized the main genetic and diet-induced animal models of NAFLD, as well as the cellular models of NAFLD. CONCLUSIONS In recent years, both the diet-induced animal models of NAFLD as well as the cellular models of NAFLD have found ever more application to investigate the mechanisms involved in NAFLD, and we referred to their advantages and disadvantages.
Collapse
Affiliation(s)
- Laura Vergani
- DISTAV, Department of Earth, Environment and Life Sciences, University of Genova, Italy
| |
Collapse
|
22
|
Di Pino A, DeFronzo RA. Insulin Resistance and Atherosclerosis: Implications for Insulin-Sensitizing Agents. Endocr Rev 2019; 40:1447-1467. [PMID: 31050706 PMCID: PMC7445419 DOI: 10.1210/er.2018-00141] [Citation(s) in RCA: 216] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 10/18/2018] [Indexed: 12/12/2022]
Abstract
Patients with type 2 diabetes mellitus (T2DM) are at high risk for macrovascular complications, which represent the major cause of mortality. Despite effective treatment of established cardiovascular (CV) risk factors (dyslipidemia, hypertension, procoagulant state), there remains a significant amount of unexplained CV risk. Insulin resistance is associated with a cluster of cardiometabolic risk factors known collectively as the insulin resistance (metabolic) syndrome (IRS). Considerable evidence, reviewed herein, suggests that insulin resistance and the IRS contribute to this unexplained CV risk in patients with T2DM. Accordingly, CV outcome trials with pioglitazone have demonstrated that this insulin-sensitizing thiazolidinedione reduces CV events in high-risk patients with T2DM. In this review the roles of insulin resistance and the IRS in the development of atherosclerotic CV disease and the impact of the insulin-sensitizing agents and of other antihyperglycemic medications on CV outcomes are discussed.
Collapse
Affiliation(s)
- Antonino Di Pino
- Diabetes Division, University of Texas Health Science Center and Texas Diabetes Institute, San Antonio, Texas
| | - Ralph A DeFronzo
- Diabetes Division, University of Texas Health Science Center and Texas Diabetes Institute, San Antonio, Texas
| |
Collapse
|
23
|
Angiotensin II Influences Pre-mRNA Splicing Regulation by Enhancing RBM20 Transcription Through Activation of the MAPK/ELK1 Signaling Pathway. Int J Mol Sci 2019; 20:ijms20205059. [PMID: 31614708 PMCID: PMC6829565 DOI: 10.3390/ijms20205059] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 10/05/2019] [Accepted: 10/07/2019] [Indexed: 12/31/2022] Open
Abstract
RNA binding motif 20 (RBM20) is a key regulator of pre-mRNA splicing of titin and other genes that are associated with cardiac diseases. Hormones, like insulin, triiodothyronine (T3), and angiotensin II (Ang II), can regulate gene-splicing through RBM20, but the detailed mechanism remains unclear. This study was aimed at investigating the signaling mechanism by which hormones regulate pre-mRNA splicing through RBM20. We first examined the role of RBM20 in Z-, I-, and M-band titin splicing at different ages in wild type (WT) and RBM20 knockout (KO) rats using RT-PCR; we found that RBM20 is the predominant regulator of I-band titin splicing at all ages. Then we treated rats with propylthiouracil (PTU), T3, streptozotocin (STZ), and Ang II and evaluated the impact of these hormones on the splicing of titin, LIM domain binding 3 (Ldb3), calcium/calmodulin-dependent protein kinase II gamma (Camk2g), and triadin (Trdn). We determined the activation of mitogen-activated protein kinase (MAPK) signaling in primary cardiomyocytes treated with insulin, T3, and Ang II using western blotting; MAPK signaling was activated and RBM20 expression increased after treatment. Two downstream transcriptional factors c-jun and ETS Transcription Factor (ELK1) can bind the promoter of RBM20. A dual-luciferase activity assay revealed that Ang II, but not insulin and T3, can trigger ELK1 and thus promote transcription of RBM20. This study revealed that Ang II can trigger ELK1 through activation of MAPK signaling by enhancing RBM20 expression which regulates pre-mRNA splicing. Our study provides a potential therapeutic target for the treatment of cardiac diseases in RBM20-mediated pre-mRNA splicing.
Collapse
|
24
|
Laskowski D, Andersson G, Humblot P, Sirard MA, Sjunnesson Y, Ferreira CR, Pirro V, Båge R. Lipid profile of bovine blastocysts exposed to insulin during in vitro oocyte maturation. Reprod Fertil Dev 2019; 30:1253-1266. [PMID: 29655403 DOI: 10.1071/rd17248] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 03/01/2018] [Indexed: 01/22/2023] Open
Abstract
Insulin is a key hormone with important functions in energy metabolism and is involved in the regulation of reproduction. Hyperinsulinaemia is known to impair fertility (for example, in obese mothers); therefore, we aimed to investigate the impact of elevated insulin concentrations during the sensitive period of oocyte maturation on gene expression and lipid profiles of the bovine Day-8 embryo. Two different insulin concentrations were used during in vitro oocyte maturation (INS10=10µgmL-1 and INS0.1=0.1µgmL-1) in order to observe possible dose-dependent effects or thresholds for hyperinsulinaemia in vitro. By investigating gene expression patterns by an mRNA microarray in combination with lipid profile analysis by desorption electrospray ionisation-mass spectrometry (DESI-MS) of embryos derived from insulin-treated oocytes, we gained further insights regarding molecular responses of embryos to insulin provocation during the first days of development. Lipid metabolism appeared to be influenced on multiple levels according to gene expression results but the profiles collected in positive-ion mode by DESI-MS (showing mostly ubiquinone, cholesteryl esters and triacylglycerols) did not differ significantly from controls. There are parallels in follicular development of ruminants and humans that make this bovine model relevant for comparative research on early human embryonic development during hyperinsulinaemia.
Collapse
Affiliation(s)
- Denise Laskowski
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, PO Box 7054, SE-750 07 Uppsala, Sweden
| | - Göran Andersson
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences, PO Box 7023, SE-750 07 Uppsala, Sweden
| | - Patrice Humblot
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, PO Box 7054, SE-750 07 Uppsala, Sweden
| | - Marc-André Sirard
- Departement des Sciences Animales, Centre de Recherche en Biologie de la Reproduction, Pavillon Des Services, Local 2732, University Laval, Québec G1V 0A6, Canada
| | - Ylva Sjunnesson
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, PO Box 7054, SE-750 07 Uppsala, Sweden
| | - Christina R Ferreira
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, IN 47907-2084, USA
| | - Valentina Pirro
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, IN 47907-2084, USA
| | - Renée Båge
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, PO Box 7054, SE-750 07 Uppsala, Sweden
| |
Collapse
|
25
|
Nseir WB, Mograbi JM, Amara AE, Abu Elheja OH, Mahamid MN. Non-alcoholic fatty liver disease and 30-day all-cause mortality in adult patients with community-acquired pneumonia. QJM 2019; 112:95-99. [PMID: 30325458 DOI: 10.1093/qjmed/hcy227] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is a common and serious form of chronic liver disease. Risk factors of NAFLD include obesity and type 2 diabetes which are associated with infections. AIM We aimed to determine the association of NAFLD with 30-day all-cause mortality in adult patients with community-acquired pneumonia (CAP). METHODS A retrospective cohort study on hospitalized patients with CAP that was conducted during a period of 4 years. We included patients aged ≥18 years with CAP who underwent abdominal ultrasonography. We compared between patients with and without NAFLD in term of age, gender, body mass index (BMI), comorbidities, CURB-65, pneumonia severity index (PSI), liver enzymes, C-reactive protein (CRP) and 30-day all-cause mortality. We used fibrosis score to distinguish between patients with NAFLD who have advanced fibrosis (F3-F4) and do not have (F0-F2). RESULTS A total of 561 patients were included in this study. The overall prevalence of NAFLD was 200/561 (35.6%). Significant differences were found between the groups with and without NAFLD in term of BMI, CURB-65, ALT, GGT and CRP. The 30-day all-cause mortality rate was 9.8% (55/561). Among the NAFLD group 34/200 (17%) subjects died vs. 21/361 (5.82%) among patients without NAFLD, P < 0.001. Multi-variate logistic regression analysis after adjusting for other multiple covariates showed that NAFLD with fibrosis score 0-2 [odds ratio (OR) 1.38, 95% confidence interval (CI) 1.12-1.51, P = 0.04], NAFLD with fibrosis score> 2 (1.52; 1.25-1.70, P = 0.03) were associated with 30-day all-cause mortality among patients with CAP. CONCLUSIONS NAFLD was associated with 30-day all-cause mortality in patients with CAP. This association was more significant in patients with advanced hepatic fibrosis.
Collapse
Affiliation(s)
- W B Nseir
- From the Division of Internal Medicine, EMMS, The Nazareth Hospital, P.O.B 8, Nazareth, Israel
- The Azrieli Faculty of Medicine, The Galilee, Bar-Ilan University, Henrietta Szold St. 8, P.O.B 1589, Safed, Israel
| | - J M Mograbi
- From the Division of Internal Medicine, EMMS, The Nazareth Hospital, P.O.B 8, Nazareth, Israel
| | - A E Amara
- From the Division of Internal Medicine, EMMS, The Nazareth Hospital, P.O.B 8, Nazareth, Israel
| | - O H Abu Elheja
- Department of Internal Medicine, Holy Family Hospital, P.O.B 11, Nazareth, Israel
| | - M N Mahamid
- From the Division of Internal Medicine, EMMS, The Nazareth Hospital, P.O.B 8, Nazareth, Israel
- The Azrieli Faculty of Medicine, The Galilee, Bar-Ilan University, Henrietta Szold St. 8, P.O.B 1589, Safed, Israel
| |
Collapse
|
26
|
Dogra S, Kar AK, Girdhar K, Daniel PV, Chatterjee S, Choubey A, Ghosh S, Patnaik S, Ghosh D, Mondal P. Zinc oxide nanoparticles attenuate hepatic steatosis development in high-fat-diet fed mice through activated AMPK signaling axis. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 17:210-222. [PMID: 30708053 DOI: 10.1016/j.nano.2019.01.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 11/22/2018] [Accepted: 01/11/2019] [Indexed: 01/01/2023]
Abstract
Insulin resistance is thought to be a common link between obesity and Non-Alcoholic Fatty Liver Disease (NAFLD). NAFLD has now reached epidemic status worldwide and identification of molecules or pathways as newer therapeutic strategies either to prevent or overcome insulin resistance seems critical. Dysregulated hepatic lipogenesis (DNL) is a hallmark of NAFLD in humans and rodents. Therefore, reducing DNL accretion may be critical in the development of therapeutics of NAFLD. In our in vivo model (high-fat-diet fed [HFD] obese mice) we found Zinc oxide nanoparticles (ZnO NPs) significantly decreased HFD-induced hepatic steatosis and peripheral insulin resistance. This protective mechanism of ZnO NPs was signaled through hepatic SIRT1-LKB1-AMPK which restricted SREBP-1c within the cytosol limiting its transcriptional ability and thereby ameliorating HFD mediated DNL. These observations indicate that ZnO NP can serve as a therapeutic strategy to improve the physiological homeostasis during obesity and its associated metabolic abnormalities.
Collapse
Affiliation(s)
- Surbhi Dogra
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, H.P, India
| | - Aditya K Kar
- CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, Uttar Pradesh, India
| | - Khyati Girdhar
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, H.P, India
| | - P Vineeth Daniel
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, H.P, India
| | - Swarup Chatterjee
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, H.P, India
| | - Abhinav Choubey
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, H.P, India
| | - Subrata Ghosh
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, H.P, India
| | - Satyakam Patnaik
- CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, Uttar Pradesh, India
| | - Debabrata Ghosh
- CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, Uttar Pradesh, India.
| | - Prosenjit Mondal
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, H.P, India.
| |
Collapse
|
27
|
Berthier A, Vinod M, Porez G, Steenackers A, Alexandre J, Yamakawa N, Gheeraert C, Ploton M, Maréchal X, Dubois-Chevalier J, Hovasse A, Schaeffer-Reiss C, Cianférani S, Rolando C, Bray F, Duez H, Eeckhoute J, Lefebvre T, Staels B, Lefebvre P. Combinatorial regulation of hepatic cytoplasmic signaling and nuclear transcriptional events by the OGT/REV-ERBα complex. Proc Natl Acad Sci U S A 2018; 115:E11033-E11042. [PMID: 30397120 PMCID: PMC6255172 DOI: 10.1073/pnas.1805397115] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The nuclear receptor REV-ERBα integrates the circadian clock with hepatic glucose and lipid metabolism by nucleating transcriptional comodulators at genomic regulatory regions. An interactomic approach identified O-GlcNAc transferase (OGT) as a REV-ERBα-interacting protein. By shielding cytoplasmic OGT from proteasomal degradation and favoring OGT activity in the nucleus, REV-ERBα cyclically increased O-GlcNAcylation of multiple cytoplasmic and nuclear proteins as a function of its rhythmically regulated expression, while REV-ERBα ligands mostly affected cytoplasmic OGT activity. We illustrate this finding by showing that REV-ERBα controls OGT-dependent activities of the cytoplasmic protein kinase AKT, an essential relay in insulin signaling, and of ten-of-eleven translocation (TET) enzymes in the nucleus. AKT phosphorylation was inversely correlated to REV-ERBα expression. REV-ERBα enhanced TET activity and DNA hydroxymethylated cytosine (5hmC) levels in the vicinity of REV-ERBα genomic binding sites. As an example, we show that the REV-ERBα/OGT complex modulates SREBP-1c gene expression throughout the fasting/feeding periods by first repressing AKT phosphorylation and by epigenomically priming the Srebf1 promoter for a further rapid response to insulin. Conclusion: REV-ERBα regulates cytoplasmic and nuclear OGT-controlled processes that integrate at the hepatic SREBF1 locus to control basal and insulin-induced expression of the temporally and nutritionally regulated lipogenic SREBP-1c transcript.
Collapse
Affiliation(s)
- Alexandre Berthier
- University of Lille, Inserm, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, European Genomic Institute for Diabetes, U1011, Lille F-59045, France
| | - Manjula Vinod
- University of Lille, Inserm, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, European Genomic Institute for Diabetes, U1011, Lille F-59045, France
| | - Geoffrey Porez
- University of Lille, Inserm, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, European Genomic Institute for Diabetes, U1011, Lille F-59045, France
| | - Agata Steenackers
- University of Lille, CNRS, Unité de Glycobiologie Structurale et Fonctionnelle, UMR 8576, Villeneuve d'Ascq F-59655, France
| | - Jérémy Alexandre
- University of Lille, Inserm, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, European Genomic Institute for Diabetes, U1011, Lille F-59045, France
| | - Nao Yamakawa
- University of Lille, CNRS, Unité de Glycobiologie Structurale et Fonctionnelle, UMR 8576, Villeneuve d'Ascq F-59655, France
| | - Céline Gheeraert
- University of Lille, Inserm, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, European Genomic Institute for Diabetes, U1011, Lille F-59045, France
| | - Maheul Ploton
- University of Lille, Inserm, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, European Genomic Institute for Diabetes, U1011, Lille F-59045, France
| | - Xavier Maréchal
- University of Lille, Inserm, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, European Genomic Institute for Diabetes, U1011, Lille F-59045, France
| | - Julie Dubois-Chevalier
- University of Lille, Inserm, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, European Genomic Institute for Diabetes, U1011, Lille F-59045, France
| | - Agnès Hovasse
- Laboratoire de Spectrométrie de Masse BioOrganique, University of Strasbourg, CNRS, Institut Pluridisciplinaire Hubert Curien, UMR 7178, Strasbourg F-67037, France
| | - Christine Schaeffer-Reiss
- Laboratoire de Spectrométrie de Masse BioOrganique, University of Strasbourg, CNRS, Institut Pluridisciplinaire Hubert Curien, UMR 7178, Strasbourg F-67037, France
| | - Sarah Cianférani
- Laboratoire de Spectrométrie de Masse BioOrganique, University of Strasbourg, CNRS, Institut Pluridisciplinaire Hubert Curien, UMR 7178, Strasbourg F-67037, France
| | - Christian Rolando
- Miniaturisation pour la Synthèse, l'Analyse & la Protéomique, CNRS, Unité de Service et de Recherche (USR) 3290, University of Lille, Villeneuve d'Ascq F-59655, France
- Fédération de Recherche Biochimie Structurale et Fonctionnelle des Assemblages Biomoléculaires FRABio, FR 3688 CNRS, University of Lille, Villeneuve d'Ascq F-59655, France
- Institut M.-E. Chevreul, CNRS, FR 2638, University of Lille, Villeneuve d'Ascq F-59655, France
| | - Fabrice Bray
- Miniaturisation pour la Synthèse, l'Analyse & la Protéomique, CNRS, Unité de Service et de Recherche (USR) 3290, University of Lille, Villeneuve d'Ascq F-59655, France
- Fédération de Recherche Biochimie Structurale et Fonctionnelle des Assemblages Biomoléculaires FRABio, FR 3688 CNRS, University of Lille, Villeneuve d'Ascq F-59655, France
- Institut M.-E. Chevreul, CNRS, FR 2638, University of Lille, Villeneuve d'Ascq F-59655, France
| | - Hélène Duez
- University of Lille, Inserm, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, European Genomic Institute for Diabetes, U1011, Lille F-59045, France
| | - Jérôme Eeckhoute
- University of Lille, Inserm, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, European Genomic Institute for Diabetes, U1011, Lille F-59045, France
| | - Tony Lefebvre
- University of Lille, CNRS, Unité de Glycobiologie Structurale et Fonctionnelle, UMR 8576, Villeneuve d'Ascq F-59655, France
| | - Bart Staels
- University of Lille, Inserm, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, European Genomic Institute for Diabetes, U1011, Lille F-59045, France
| | - Philippe Lefebvre
- University of Lille, Inserm, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, European Genomic Institute for Diabetes, U1011, Lille F-59045, France;
| |
Collapse
|
28
|
Howell GE, McDevitt E, Henein L, Mulligan C, Young D. Alterations in cellular lipid metabolism produce neutral lipid accumulation following exposure to the organochlorine compound trans-nonachlor in rat primary hepatocytes. ENVIRONMENTAL TOXICOLOGY 2018; 33:962-971. [PMID: 29964320 PMCID: PMC6105551 DOI: 10.1002/tox.22583] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 05/21/2018] [Accepted: 05/28/2018] [Indexed: 05/16/2023]
Abstract
Recent epidemiological studies have revealed significant positive associations between exposure to organochlorine (OC) pesticides and occurrence of the metabolic syndrome and there are a growing number of animal-based studies to support causality. However, the cellular mechanisms linking OC compound exposure and metabolic dysfunction remain elusive. Therefore, the present study was designed to determine if direct exposure to three highly implicated OC compounds promoted hepatic steatosis, the hepatic ramification of the metabolic syndrome. First, the steatotic effect of p,p'-dichlorodiphenyldichloroethylene (DDE), oxychlordane, and trans-nonachlor was determined in freshly isolated rat primary hepatocytes. Exposure to trans-nonachlor significantly increased neutral lipid accumulation as opposed to DDE and oxychlordane. To determine possible mechanisms governing increased fatty acid availability, the effects of trans-nonachlor exposure on fatty acid uptake, de novo lipogenesis, triglyceride secretion, and fatty acid oxidation were explored. Trans-nonachlor did not significantly alter fatty acid uptake. However, insulin-stimulated de novo lipogenesis as well as basal expression of fatty acid synthase, a major regulator of lipogenesis were significantly increased following trans-nonachlor exposure. Interestingly, there was a significant decrease in fatty acid oxidation following trans-nonachlor exposure. This decrease in fatty acid oxidation was accompanied by a slight, but significant increase in oleic acid-induced cellular triglyceride secretion. Therefore, taken together, the present data indicate direct exposure to trans-nonachlor has a more potent pro-steatotic effect than exposure to DDE or oxychlordane. This pro-steatotic effect of trans-nonachlor appears to be predominately mediated via increased de novo lipogenesis and decreased fatty acid oxidation.
Collapse
Affiliation(s)
- George Eli Howell
- Mississippi State University College of Veterinary Medicine, Department of Basic Sciences, Mississippi State, MS 39762
- Corresponding author: George Eli Howell III, Ph.D, 240 Wise Center Drive, P.O. Box 6100, Mississippi State, MS 39762, Phone: 601-420-4707, Fax: 662-325-1031,
| | - Erin McDevitt
- Mississippi State University College of Veterinary Medicine, Department of Basic Sciences, Mississippi State, MS 39762
| | - Lucie Henein
- Mississippi State University College of Veterinary Medicine, Department of Basic Sciences, Mississippi State, MS 39762
| | - Charlee Mulligan
- Mississippi State University College of Veterinary Medicine, Department of Basic Sciences, Mississippi State, MS 39762
| | - Darian Young
- Mississippi State University College of Veterinary Medicine, Department of Basic Sciences, Mississippi State, MS 39762
| |
Collapse
|
29
|
Elkahoui S, Bartley GE, Yokoyama WH, Friedman M. Dietary Supplementation of Potato Peel Powders Prepared from Conventional and Organic Russet and Non-organic Gold and Red Potatoes Reduces Weight Gain in Mice on a High-Fat Diet. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:6064-6072. [PMID: 29877090 DOI: 10.1021/acs.jafc.8b01987] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The present study investigated the potential of potato peel powders, high in bioactive phenolic compounds and glycoalkaloids, to reduce weight gain in mice consuming a high-fat diet. Potato peel powders were prepared from the following fresh commercial potato varieties by hand-peeling and then freeze-drying and grinding the peels into powder: non-organic (conventionally grown) gold, red, and Russet and organically grown Russet. Mice diets (25% fat by weight) were supplemented with either 10 or 20% potato peel powders for 3 weeks. In comparison to the control diet, the isocaloric and isonitrogenous peel-containing diets induced a reduction in weight gain that ranged from 17-45% (10% peel diets) to 46-73% (20% peel diets), suggesting that differences in weight gain are associated with the potato peel source and peel concentration of the diet. Weight reductions were accompanied by reduced epididymal white adipose tissue ranging from 22 to 80% as well as changes in the microbiota analyzed using next-generation sequencing and in obesity-associated genetic biomarkers determined by the quantitative real-time polymerase chain reaction. Safety aspects and possible mechanisms of the antiobesity effects are discussed in terms of the composition of the bioactive potato peel compounds, which were determined using high-performance liquid chromatography. The results suggest that potato peels, a major peeling byproduct of potato processing used to prepare fries, chips, and potato flour, that showed exceptionally high antiobesity properties in fat mice, have the potential to serve as an antiobesity functional food.
Collapse
Affiliation(s)
- Salem Elkahoui
- Laboratory of Bioactive Substances , Center of Biotechnology of Borj Cédria , BP 901, 2050 Hammam-Lif , Tunisia
| | - Glenn E Bartley
- Healthy Processed Foods Research, Western Regional Research Center, Agricultural Research Service , United States Department of Agriculture , Albany , California 94710 , United States
| | - Wallace H Yokoyama
- Healthy Processed Foods Research, Western Regional Research Center, Agricultural Research Service , United States Department of Agriculture , Albany , California 94710 , United States
| | - Mendel Friedman
- Healthy Processed Foods Research, Western Regional Research Center, Agricultural Research Service , United States Department of Agriculture , Albany , California 94710 , United States
| |
Collapse
|
30
|
The utility of noninvasive scores in assessing the prevalence of nonalcoholic fatty liver disease and advanced fibrosis in type 1 diabetic patients. Hepatol Int 2018; 12:37-43. [PMID: 29318450 DOI: 10.1007/s12072-017-9840-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 12/25/2017] [Indexed: 12/18/2022]
Abstract
GOALS The aim of our study is to assess the prevalence of nonalcoholic fatty liver disease (NAFLD) and advanced hepatic fibrosis in patients with type 1 diabetes (T1D) using simple noninvasive scores. BACKGROUND There is paucity of data on the prevalence of NAFLD in T1D. Moreover, T1D could be a risk factor for advanced disease in NAFLD patients. STUDY Using ICD-9 codes, all patients with the diagnosis of T1D were reviewed and a retrospective chart analysis was carried out on 23,314 patients between the ages of 18 and 80. To predict the prevalence of NAFLD, we calculated the hepatic steatosis index (HSI). To estimate the prevalence of advanced fibrosis, NAFLD fibrosis score (NFS), FIB-4 index, AST to platelet ratio index (APRI), and AST/ALT ratio were calculated. RESULTS Of the 4899 patients included in the analysis, 86.9% were Caucasian and 67% were above normal weight limit. NAFLD based on HSI > 36 was present in 71.3% of patients. Advanced fibrosis was present in 20.3% based on NFS > 0.676, 6.7% based on FIB-4 > 2.67, 2.1% based on APRI > 1.5, and 22.1% based on AST/ALT > 1.4%, indicating a high risk of developing cirrhosis and end-stage liver disease. CONCLUSION In this large cohort of patients with T1DM, we detected a high prevalence of hepatic steatosis and advanced fibrosis using noninvasive scores. These scores are easy and inexpensive tools to screen for NAFLD and advanced fibrosis, although the significant variability of the percentage of advanced fibrosis using these scores indicates the need for further validation in diabetic populations. CLINICAL TRIAL REGISTRATION NUMBER CCF-16-018.
Collapse
|
31
|
Zheng ZG, Lu C, Thu PM, Zhang X, Li HJ, Li P, Xu X. Praeruptorin B improves diet-induced hyperlipidemia and alleviates insulin resistance via regulating SREBP signaling pathway. RSC Adv 2018. [DOI: 10.1039/c7ra11797c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Many metabolic diseases are caused by disruption of lipid homeostasis.
Collapse
Affiliation(s)
- Zu-Guo Zheng
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing
- China
| | - Chong Lu
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing
- China
| | - Pyone Myat Thu
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing
- China
| | - Xin Zhang
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing
- China
| | - Hui-Jun Li
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing
- China
| | - Ping Li
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing
- China
| | - Xiaojun Xu
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing
- China
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases
| |
Collapse
|
32
|
Huang LH, Chung HY, Su HM. Docosahexaenoic acid reduces sterol regulatory element binding protein-1 and fatty acid synthase expression and inhibits cell proliferation by inhibiting pAkt signaling in a human breast cancer MCF-7 cell line. BMC Cancer 2017; 17:890. [PMID: 29282029 PMCID: PMC5745739 DOI: 10.1186/s12885-017-3936-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 12/19/2017] [Indexed: 12/02/2022] Open
Abstract
Background Fatty acid synthase (FASN), the major enzyme in de novo fatty acid synthesis, is highly expressed in breast cancer and its expression is reduced by polyunsaturated fatty acids (PUFAs) in liver. We previously found a positive association between rat mammary tumor levels of the n-6 PUFA arachidonic acid (AA) and tumor weight. We examined the roles of the major n-3 PUFA, docosahexaenoic acid (DHA, 22:6n-3), and the major n-6 PUFA, AA, in FASN expression in, and proliferation of, human breast cancer MCF-7 cells. Methods The cells were treated for 48 h with BSA or 60 μM BSA-bound DHA, AA, or oleic acid (OA, 18:1n-9), then were incubated with or without estradiol or insulin. Western blot and 3H–thymidine incorporation assay were used to determine the role of DHA on FASN regulation and MCF-7 cell proliferation. Results DHA, but neither AA nor OA, inhibits estradiol-induced and insulin-induced expression of the precursor of sterol regulatory element binding protein-1 (p-SREBP-1), its mature form (m-SREBP-1), and FASN. Estradiol or insulin stimulation increased the pAkt/Akt and pS6/S6 ratios, expression of p-SREBP-1, m-SREBP-1, and FASN, and cell proliferation, and these effects were decreased by DHA. The DHA-induced decrease in FASN expression resulted from reduced pAkt/Akt signaling and not pERK1/2/ERK1/2 signaling. In addition, DHA enhanced the inhibitory effect of LY294002 on pAkt signaling and expression of p-SREBP-1, m-SREBP-1, and FASN. However, addition of rapamycin, an inhibitor of the mTOR signaling pathways, 1 h before addition of estradiol or insulin increased the pAkt/Akt ratio and FASN expression, and this effect was inhibited by addition of DHA 48 h before rapamycin. Conclusion We conclude that, in MCF-7 cells, DHA inhibits pAKT signaling and thus expression of p-SREBP-1, m-SREBP-1, and FASN and cell proliferation.
Collapse
Affiliation(s)
- Li-Hsuan Huang
- Institute of Physiology, College of Medicine, National Taiwan University, 1 Sec 1 Jai-Ai Rd, Taipei, 100, Taiwan
| | - Hsin-Yun Chung
- Institute of Physiology, College of Medicine, National Taiwan University, 1 Sec 1 Jai-Ai Rd, Taipei, 100, Taiwan
| | - Hui-Min Su
- Institute of Physiology, College of Medicine, National Taiwan University, 1 Sec 1 Jai-Ai Rd, Taipei, 100, Taiwan.
| |
Collapse
|
33
|
Insulin Resistance and NAFLD: A Dangerous Liaison beyond the Genetics. CHILDREN-BASEL 2017; 4:children4080074. [PMID: 28805745 PMCID: PMC5575596 DOI: 10.3390/children4080074] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 08/02/2017] [Accepted: 08/03/2017] [Indexed: 02/08/2023]
Abstract
Over the last decade, the understanding of the association between insulin resistance (IR) and non-alcoholic fatty liver disease (NAFLD) has dramatically evolved. There is clear understanding that carriers of some common genetic variants, i.e., the patatin-like phospholipase domain-containing 3 (PNPLA3) or the transmembrane 6 superfamily member 2 (TM6SF2) are at risk of developing severe forms of NAFLD even in the presence of reduced or absent IR. In contrast, there are obese patients with “metabolic” (non-genetically driven) NAFLD who present severe IR. Owing to the epidemic obesity and the high prevalence of these genetic variants in the general population, the number of pediatric cases with combination of genetic and metabolic NAFLD is expected to be very high. Gut dysbiosis, excessive dietary intake of saturated fats/fructose-enriched foods and exposure to some chemicals contribute all to both IR and NAFLD, adding further complexity to the understanding of their relationship. Once NAFLD is established, IR can accelerate the progression to the more severe form of liver derangement that is the non-alcoholic steatohepatitis.
Collapse
|
34
|
Taguchi Y, Toyoshima Y, Tokita R, Kato H, Takahashi SI, Minami S. Triglyceride synthesis in hepatocytes isolated from rats fed a low-protein diet is enhanced independently of upregulation of insulin signaling. Biochem Biophys Res Commun 2017. [DOI: 10.1016/j.bbrc.2017.06.120] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
35
|
Momtazi AA, Banach M, Pirro M, Stein EA, Sahebkar A. PCSK9 and diabetes: is there a link? Drug Discov Today 2017; 22:883-895. [DOI: 10.1016/j.drudis.2017.01.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 12/08/2016] [Accepted: 01/10/2017] [Indexed: 12/14/2022]
|
36
|
Xie Z, Wan X, Zhong L, Yang H, Li P, Xu X. Carnosic acid alleviates hyperlipidemia and insulin resistance by promoting the degradation of SREBPs via the 26S proteasome. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.01.040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
37
|
So WY, Leung PS. Irisin ameliorates hepatic glucose/lipid metabolism and enhances cell survival in insulin-resistant human HepG2 cells through adenosine monophosphate-activated protein kinase signaling. Int J Biochem Cell Biol 2016; 78:237-247. [PMID: 27452313 DOI: 10.1016/j.biocel.2016.07.022] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 07/18/2016] [Accepted: 07/20/2016] [Indexed: 02/06/2023]
Abstract
Irisin is a newly identified myokine that promotes the browning of white adipose tissue, enhances glucose uptake in skeletal muscle and modulates hepatic metabolism. However, the signaling pathways involved in the effects on hepatic glucose and lipid metabolism have not been resolved. This study aimed to examine the role of irisin in the regulation of hepatic glucose/lipid metabolism and cell survival, and whether adenosine monophosphate-activated protein kinase (AMPK), a master metabolic regulator in the liver, is involved in irisin's actions. Human liver-derived HepG2 cells were cultured in normal glucose-normal insulin (NGNI) or high glucose-high insulin (HGHI/insulin-resistant) condition. Hepatic glucose and lipid metabolism was evaluated by glucose output and glycogen content or triglyceride accumulation assays, respectively. Our results showed that irisin stimulated phosphorylation of AMPK and acetyl-CoA-carboxylase (ACC) via liver kinase B1 (LKB1) rather than Ca(2+)/calmodulin-dependent protein kinase kinase β (CaMKKβ) in HepG2 cells. Irisin ameliorated hepatic insulin resistance induced by HGHI condition. Irisin reduced hepatic triglyceride content and glucose output, but increased glycogen content, with those effects reversed by dorsomorphin, an AMPK inhibitor. Furthermore, irisin also stimulated extracellular signal-regulated kinase (ERK) 1/2 phosphorylation and promoted cell survival in an AMPK-dependent manner. In conclusion, our data indicate that irisin ameliorates dysregulation of hepatic glucose/lipid metabolism and cell death in insulin-resistant states via AMPK activation. These findings reveal a novel irisin-mediated protective mechanism in hepatic metabolism which provides a scientific basis for irisin as a potential therapeutic target for the treatment of insulin resistance and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Wing Yan So
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.
| | - Po Sing Leung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
38
|
Karahashi M, Hirata-Hanta Y, Kawabata K, Tsutsumi D, Kametani M, Takamatsu N, Sakamoto T, Yamazaki T, Asano S, Mitsumoto A, Kawashima Y, Kudo N. Abnormalities in the Metabolism of Fatty Acids and Triacylglycerols in the Liver of the Goto-Kakizaki Rat: A Model for Non-Obese Type 2 Diabetes. Lipids 2016; 51:955-71. [PMID: 27372943 DOI: 10.1007/s11745-016-4171-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 06/20/2016] [Indexed: 02/08/2023]
Abstract
The Goto-Kakizaki (GK) rat is widely used as an animal model for spontaneous-onset type 2 diabetes without obesity; nevertheless, little information is available on the metabolism of fatty acids and triacylglycerols (TAG) in their livers. We investigated the mechanisms underlying the alterations in the metabolism of fatty acids and TAG in their livers, in comparison with Zucker (fa/fa) rats, which are obese and insulin resistant. Lipid profiles, the expression of genes for enzymes and proteins related to the metabolism of fatty acid and TAG, de novo synthesis of fatty acids and TAG in vivo, fatty acid synthase activity in vitro, fatty acid oxidation in liver slices, and very-low-density-lipoprotein (VLDL)-TAG secretion in vivo were estimated. Our results revealed that (1) the TAG accumulation was moderate, (2) the de novo fatty acid synthesis was increased by upregulation of fatty acid synthase in a post-transcriptional manner, (3) fatty acid oxidation was also augmented through the induction of carnitine palmitoyltransferase 1a, and (4) the secretion rate of VLDL-TAG remained unchanged in the livers of GK rats. These results suggest that, despite the fact that GK rats exhibit non-obese type 2 diabetes, the upregulation of de novo lipogenesis is largely compensated by the upregulation of fatty acid oxidation, resulting in only moderate increase in TAG accumulation in the liver.
Collapse
Affiliation(s)
- Minako Karahashi
- School of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan
| | - Yuko Hirata-Hanta
- School of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan
| | - Kohei Kawabata
- School of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan
| | - Daisuke Tsutsumi
- School of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan
| | - Misaki Kametani
- School of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan
| | - Nanako Takamatsu
- School of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan
| | - Takeshi Sakamoto
- School of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan
| | - Tohru Yamazaki
- School of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan
| | - Satoshi Asano
- Department of Pharmaceutical Sciences, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara, Tochigi, 324-8501, Japan
| | - Atsushi Mitsumoto
- Faculty of Pharmaceutical Sciences, Josai International University, 1 Gumyo, Togane, Chiba, 283-8555, Japan
| | - Yoichi Kawashima
- School of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan
| | - Naomi Kudo
- School of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan.
| |
Collapse
|
39
|
Regnault C, Willison J, Veyrenc S, Airieau A, Méresse P, Fortier M, Fournier M, Brousseau P, Raveton M, Reynaud S. Metabolic and immune impairments induced by the endocrine disruptors benzo[a]pyrene and triclosan in Xenopus tropicalis. CHEMOSPHERE 2016; 155:519-527. [PMID: 27153234 DOI: 10.1016/j.chemosphere.2016.04.047] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 04/09/2016] [Accepted: 04/12/2016] [Indexed: 06/05/2023]
Abstract
Despite numerous studies suggesting that amphibians are highly sensitive to cumulative anthropogenic stresses, the role played by endocrine disruptors (EDs) in the decline of amphibian populations remains unclear. EDs have been extensively studied in adult amphibians for their capacity to disturb reproduction by interfering with the sexual hormone axis. Here, we studied the in vivo responses of Xenopus tropicalis males exposed to environmentally relevant concentrations of each ED, benzo[a]pyrene (BaP) and triclosan (TCS) alone (10 μg L(-1)) or a mixture of the two (10 μg L(-1) each) over a 24 h exposure period by following the modulation of the transcription of key genes involved in metabolic, sexual and immunity processes and the cellular changes in liver, spleen and testis. BaP, TCS and the mixture of the two all induced a marked metabolic disorder in the liver highlighted by insulin resistance-like and non-alcoholic fatty liver disease (NAFLD)-like phenotypes together with hepatotoxicity due to the impairment of lipid metabolism. For TCS and the mixture, these metabolic disorders were concomitant with modulation of innate immunity. These results confirmed that in addition to the reproductive effects induced by EDs in amphibians, metabolic disorders and immune system disruption should also be considered.
Collapse
Affiliation(s)
- Christophe Regnault
- Univ. Grenoble-Alpes, LECA, F-38000, Grenoble, France; CNRS, LECA, F-38000, Grenoble, France; Univ. Grenoble-Alpes, BEeSy, F-38000, Grenoble, France.
| | - John Willison
- Univ. Grenoble-Alpes, Institut de recherches en technologies et Sciences pour le vivant, Laboratoire de chimie et biologie des métaux (iRTSV-LCBM), F-38000, France; CNRS, IRTSV-LCBM, F-38000, Grenoble, France; Commissariat à l'énergie atomique et aux énergies alternatives (CEA), iRTSV-LCBM, F-38000, Grenoble, France.
| | - Sylvie Veyrenc
- Univ. Grenoble-Alpes, LECA, F-38000, Grenoble, France; CNRS, LECA, F-38000, Grenoble, France; Univ. Grenoble-Alpes, BEeSy, F-38000, Grenoble, France.
| | - Antinéa Airieau
- Univ. Grenoble-Alpes, LECA, F-38000, Grenoble, France; CNRS, LECA, F-38000, Grenoble, France; Univ. Grenoble-Alpes, BEeSy, F-38000, Grenoble, France.
| | - Patrick Méresse
- Univ. Grenoble-Alpes, LECA, F-38000, Grenoble, France; Univ. Grenoble-Alpes, CUBE, F-38000, Grenoble, France.
| | | | | | | | - Muriel Raveton
- Univ. Grenoble-Alpes, LECA, F-38000, Grenoble, France; CNRS, LECA, F-38000, Grenoble, France; Univ. Grenoble-Alpes, BEeSy, F-38000, Grenoble, France.
| | - Stéphane Reynaud
- Univ. Grenoble-Alpes, LECA, F-38000, Grenoble, France; CNRS, LECA, F-38000, Grenoble, France; Univ. Grenoble-Alpes, BEeSy, F-38000, Grenoble, France.
| |
Collapse
|
40
|
Klimentidis YC, Arora A. Interaction of Insulin Resistance and Related Genetic Variants With Triglyceride-Associated Genetic Variants. ACTA ACUST UNITED AC 2016; 9:154-61. [PMID: 26850992 DOI: 10.1161/circgenetics.115.001246] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 01/27/2016] [Indexed: 12/24/2022]
Abstract
BACKGROUND Several studies suggest that some triglyceride-associated single-nucleotide polymorphisms (SNPs) have pleiotropic and opposite effects on glycemic traits. This potentially implicates them in pathways such as de novo lipogenesis, which is presumably upregulated in the context of insulin resistance. We therefore tested whether the association of triglyceride-associated SNPs with triglyceride levels differs according to one's level of insulin resistance. METHODS AND RESULTS In 3 cohort studies (combined n=12 487), we tested the interaction of established triglyceride-associated SNPs (individually and collectively) with several traits related to insulin resistance, on triglyceride levels. We also tested the interaction of triglyceride SNPs with fasting insulin-associated SNPs, individually and collectively, on triglyceride levels. We find significant interactions of a weighted genetic risk score for triglycerides with insulin resistance on triglyceride levels (Pinteraction=2.73×10(-11) and Pinteraction=2.48×10(-11) for fasting insulin and homeostasis model assessment of insulin resistance, respectively). The association of the triglyceride genetic risk score with triglyceride levels is >60% stronger among those in the highest tertile of homeostasis model assessment of insulin resistance compared with those in the lowest tertile. Individual SNPs contributing to this trend include those in/near GCKR, CILP2, and IRS1, whereas PIGV-NROB2 and LRPAP1 display an opposite trend of interaction. In the pooled data set, we also identify a SNP-by-SNP interaction involving a triglyceride-associated SNP, rs4722551 near MIR148A, with a fasting insulin-associated SNP, rs4865796 in ARL15 (Pinteraction=4.1×10(-5)). CONCLUSIONS Our findings may thus provide genetic evidence for the upregulation of triglyceride levels in insulin-resistant individuals, in addition to identifying specific genetic loci and a SNP-by-SNP interaction implicated in this process.
Collapse
Affiliation(s)
- Yann C Klimentidis
- From the Department of Epidemiology and Biostatistics, Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson.
| | - Amit Arora
- From the Department of Epidemiology and Biostatistics, Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson
| |
Collapse
|
41
|
Yuan L, Bambha K. Bile acid receptors and nonalcoholic fatty liver disease. World J Hepatol 2015; 7:2811-2818. [PMID: 26668692 PMCID: PMC4670952 DOI: 10.4254/wjh.v7.i28.2811] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 09/14/2015] [Accepted: 11/25/2015] [Indexed: 02/06/2023] Open
Abstract
With the high prevalence of obesity, diabetes, and other features of the metabolic syndrome in United States, nonalcoholic fatty liver disease (NAFLD) has inevitably become a very prevalent chronic liver disease and is now emerging as one of the leading indications for liver transplantation. Insulin resistance and derangement of lipid metabolism, accompanied by activation of the pro-inflammatory response and fibrogenesis, are essential pathways in the development of the more clinically significant form of NAFLD, known as nonalcoholic steatohepatitis (NASH). Recent advances in the functional characterization of bile acid receptors, such as farnesoid X receptor (FXR) and transmembrane G protein-coupled receptor (TGR) 5, have provided further insight in the pathophysiology of NASH and have led to the development of potential therapeutic targets for NAFLD and NASH. Beyond maintaining bile acid metabolism, FXR and TGR5 also regulate lipid metabolism, maintain glucose homeostasis, increase energy expenditure, and ameliorate hepatic inflammation. These intriguing features have been exploited to develop bile acid analogues to target pathways in NAFLD and NASH pathogenesis. This review provides a brief overview of the pathogenesis of NAFLD and NASH, and then delves into the biological functions of bile acid receptors, particularly with respect to NASH pathogenesis, with a description of the associated experimental data, and, finally, we discuss the prospects of bile acid analogues in the treatment of NAFLD and NASH.
Collapse
|
42
|
Mellor CL, Steinmetz FP, Cronin MTD. The identification of nuclear receptors associated with hepatic steatosis to develop and extend adverse outcome pathways. Crit Rev Toxicol 2015; 46:138-52. [PMID: 26451809 DOI: 10.3109/10408444.2015.1089471] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The development of adverse outcome pathways (AOPs) is becoming a key component of twenty-first century toxicology. AOPs provide a conceptual framework that links the molecular initiating event to an adverse outcome through organized toxicological knowledge, bridging the gap from chemistry to toxicological effect. As nuclear receptors (NRs) play essential roles for many physiological processes within the body, they are used regularly as drug targets for therapies to treat many diseases including diabetes, cancer and neurodegenerative diseases. Due to the heightened development of NR ligands, there is increased need for the identification of related AOPs to facilitate their risk assessment. Many NR ligands have been linked specifically to steatosis. This article reviews and summarizes the role of NR and their importance with links between NR examined to identify plausible putative AOPs. The following NRs are shown to induce hepatic steatosis upon ligand binding: aryl hydrocarbon receptor, constitutive androstane receptor, oestrogen receptor, glucocorticoid receptor, farnesoid X receptor, liver X receptor, peroxisome proliferator-activated receptor, pregnane X receptor and the retinoic acid receptor. A preliminary, putative AOP was formed for NR binding linked to hepatic steatosis as the adverse outcome.
Collapse
Affiliation(s)
- Claire L Mellor
- a School of Pharmacy and Biomolecular Sciences , Liverpool John Moores University , Liverpool , England
| | - Fabian P Steinmetz
- a School of Pharmacy and Biomolecular Sciences , Liverpool John Moores University , Liverpool , England
| | - Mark T D Cronin
- a School of Pharmacy and Biomolecular Sciences , Liverpool John Moores University , Liverpool , England
| |
Collapse
|
43
|
Ferrannini E, DeFronzo RA. Impact of glucose-lowering drugs on cardiovascular disease in type 2 diabetes. Eur Heart J 2015; 36:2288-96. [PMID: 26063450 DOI: 10.1093/eurheartj/ehv239] [Citation(s) in RCA: 182] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 05/16/2015] [Indexed: 12/11/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is characterized by multiple pathophysiologic abnormalities. With time, multiple glucose-lowering medications are commonly required to reduce and maintain plasma glucose concentrations within the normal range. Type 2 diabetes mellitus individuals also are at a very high risk for microvascular complications and the incidence of heart attack and stroke is increased two- to three-fold compared with non-diabetic individuals. Therefore, when selecting medications to normalize glucose levels in T2DM patients, it is important that the agent not aggravate, and ideally even improve, cardiovascular risk factors (CVRFs) and reduce cardiovascular morbidity and mortality. In this review, we examine the effect of oral (metformin, sulfonylureas, meglitinides, thiazolidinediones, DPP4 inhibitors, SGLT2 inhibitors, and α-glucosidase inhibitors) and injectable (glucagon-like peptide-1 receptor agonists and insulin) glucose-lowering drugs on established CVRFs and long-term studies of cardiovascular outcomes. Firm evidence that in T2DM cardiovascular disease can be reversed or prevented by improving glycaemic control is still incomplete and must await large, long-term clinical trials in patients at low risk using modern treatment strategies, i.e., drug combinations designed to maximize HbA1c reduction while minimizing hypoglycaemia and excessive weight gain.
Collapse
Affiliation(s)
- Ele Ferrannini
- Institute of Clinical Physiology, National Research Council (CNR), Pisa, Italy
| | - Ralph A DeFronzo
- Diabetes Division, University of Texas Health Science Center, San Antonio, TX, USA
| |
Collapse
|
44
|
López-Soldado I, Zafra D, Duran J, Adrover A, Calbó J, Guinovart JJ. Liver glycogen reduces food intake and attenuates obesity in a high-fat diet-fed mouse model. Diabetes 2015; 64:796-807. [PMID: 25277398 DOI: 10.2337/db14-0728] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
We generated mice that overexpress protein targeting to glycogen (PTG) in the liver (PTG(OE)), which results in an increase in liver glycogen. When fed a high-fat diet (HFD), these animals reduced their food intake. The resulting effect was a lower body weight, decreased fat mass, and reduced leptin levels. Furthermore, PTG overexpression reversed the glucose intolerance and hyperinsulinemia caused by the HFD and protected against HFD-induced hepatic steatosis. Of note, when fed an HFD, PTG(OE) mice did not show the decrease in hepatic ATP content observed in control animals and had lower expression of neuropeptide Y and higher expression of proopiomelanocortin in the hypothalamus. Additionally, after an overnight fast, PTG(OE) animals presented high liver glycogen content, lower liver triacylglycerol content, and lower serum concentrations of fatty acids and β-hydroxybutyrate than control mice, regardless of whether they were fed an HFD or a standard diet. In conclusion, liver glycogen accumulation caused a reduced food intake, protected against the deleterious effects of an HFD, and diminished the metabolic impact of fasting. Therefore, we propose that hepatic glycogen content be considered a potential target for the pharmacological manipulation of diabetes and obesity.
Collapse
Affiliation(s)
- Iliana López-Soldado
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain CIBERDEM, Madrid, Spain
| | - Delia Zafra
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain CIBERDEM, Madrid, Spain
| | - Jordi Duran
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain CIBERDEM, Madrid, Spain
| | - Anna Adrover
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain
| | - Joaquim Calbó
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain CIBERDEM, Madrid, Spain
| | - Joan J Guinovart
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain CIBERDEM, Madrid, Spain Department of Biochemistry and Molecular Biology, University of Barcelona, Barcelona, Spain
| |
Collapse
|
45
|
Abstract
Accumulation of triacylglycerols within the cytoplasm of hepatocytes to the degree that lipid droplets are visible microscopically is called liver steatosis. Most commonly, it occurs when there is an imbalance between the delivery or synthesis of fatty acids in the liver and their disposal through oxidative pathways or secretion into the blood as a component of triacylglycerols in very low density lipoprotein. This disorder is called nonalcoholic fatty liver disease (NAFLD) in the absence of alcoholic abuse and viral hepatitis, and it is often associated with insulin resistance, obesity and type 2 diabetes. Also, liver steatosis can be induced by many other causes including excessive alcohol consumption, infection with genotype 3 hepatitis C virus and certain medications. Whereas hepatic triacylglycerol accumulation was once considered the ultimate effector of hepatic lipotoxicity, triacylglycerols per se are quite inert and do not induce insulin resistance or cellular injury. Rather, lipotoxic injury in the liver appears to be mediated by the global ongoing fatty acid enrichment in the liver, paralleling the development of insulin resistance. A considerable number of fatty acid metabolites may be responsible for hepatic lipotoxicity and liver injury. Additional key contributors include hepatic cytosolic lipases and the "lipophagy" of lipid droplets, as sources of hepatic fatty acids. The specific origin of the lipids, mainly triacylglycerols, accumulating in liver has been unraveled by recent kinetic studies, and identifying the origin of the accumulated triacylglycerols in the liver of patients with NAFLD may direct the prevention and treatment of this condition.
Collapse
Affiliation(s)
- David Q-H Wang
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Saint Louis University School of Medicine, St. Louis, Missouri
| | | | | |
Collapse
|
46
|
Zhuo MQ, Luo Z, Wu K, Zhu QL, Zheng JL, Zhang LH, Chen QL. Regulation of insulin on lipid metabolism in freshly isolated hepatocytes from yellow catfish (Pelteobagrus fulvidraco). Comp Biochem Physiol B Biochem Mol Biol 2014; 177-178:21-8. [DOI: 10.1016/j.cbpb.2014.08.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 07/31/2014] [Accepted: 08/06/2014] [Indexed: 12/12/2022]
|
47
|
Laggai S, Kessler SM, Boettcher S, Lebrun V, Gemperlein K, Lederer E, Leclercq IA, Mueller R, Hartmann RW, Haybaeck J, Kiemer AK. The IGF2 mRNA binding protein p62/IGF2BP2-2 induces fatty acid elongation as a critical feature of steatosis. J Lipid Res 2014; 55:1087-97. [PMID: 24755648 DOI: 10.1194/jlr.m045500] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Indexed: 12/12/2022] Open
Abstract
Liver-specific overexpression of the insulin-like growth factor 2 (IGF2) mRNA binding protein p62/IGF2BP2-2 induces a fatty liver, which highly expresses IGF2 Because IGF2 expression is elevated in patients with steatohepatitis, the aim of our study was to elucidate the role and interconnection of p62 and IGF2 in lipid metabolism. Expression of p62 and IGF2 highly correlated in human liver disease. p62 induced an elevated ratio of C18:C16 and increased fatty acid elongase 6 (ELOVL6) protein, the enzyme catalyzing the elongation of C16 to C18 fatty acids and promoting nonalcoholic steatohepatitis in mice and humans. The p62 overexpression induced the activation of the ELOVL6 transcriptional activator sterol regulatory element binding transcription factor 1 (SREBF1). Recombinant IGF2 induced the nuclear translocation of SREBF1 and a neutralizing IGF2 antibody reduced ELOVL6 and mature SREBF1 protein levels. Concordantly, p62 and IGF2 correlated with ELOVL6 in human livers. Decreased palmitoyl-CoA levels, as found in p62 transgenic livers, can explain the lipogenic action of ELOVL6. Accordingly, p62 represents an inducer of hepatic C18 fatty acid production via a SREBF1-dependent induction of ELOVL6. These findings underline the detrimental role of p62 in liver disease.
Collapse
Affiliation(s)
- Stephan Laggai
- Department of Pharmacy, Pharmaceutical Biology, Department of Pharmacy, Pharmaceutical, Saarland University, Saarbrücken, Germany
| | - Sonja M Kessler
- Department of Pharmacy, Pharmaceutical Biology, Department of Pharmacy, Pharmaceutical, Saarland University, Saarbrücken, Germany Medicinal Chemistry, Saarland University, Saarbrücken, Germany Laboratory of Hepato-gastroenterology, Institut de Recherche expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | | | - Valérie Lebrun
- Laboratory of Hepato-gastroenterology, Institut de Recherche expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | - Katja Gemperlein
- Department of Pharmacy, Pharmaceutical Biotechnology, Saarland University, Saarbrücken, Germany Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarbrücken, Germany
| | - Eva Lederer
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Isabelle A Leclercq
- Laboratory of Hepato-gastroenterology, Institut de Recherche expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | - Rolf Mueller
- Department of Pharmacy, Pharmaceutical Biotechnology, Saarland University, Saarbrücken, Germany Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarbrücken, Germany
| | - Rolf W Hartmann
- Medicinal Chemistry, Saarland University, Saarbrücken, Germany Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarbrücken, Germany
| | | | - Alexandra K Kiemer
- Department of Pharmacy, Pharmaceutical Biology, Department of Pharmacy, Pharmaceutical, Saarland University, Saarbrücken, Germany
| |
Collapse
|
48
|
Cilostazol inhibits insulin-stimulated expression of sterol regulatory binding protein-1c via inhibition of LXR and Sp1. Exp Mol Med 2014; 46:e73. [PMID: 24458133 PMCID: PMC3909891 DOI: 10.1038/emm.2013.143] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 10/10/2013] [Accepted: 10/21/2013] [Indexed: 12/20/2022] Open
Abstract
Hepatic steatosis is common in obese individuals with hyperinsulinemia and is an important hepatic manifestation of metabolic syndrome. Sterol regulatory binding protein-1c (SREBP-1c) is a master regulator of lipogenic gene expression in the liver. Hyperinsulinemia induces transcription of SREBP-1c via activation of liver X receptor (LXR) and specificity protein 1 (Sp1). Cilostazol is an antiplatelet agent that prevents atherosclerosis and decreases serum triglyceride levels. However, little is known about the effects of cilostazol on hepatic lipogenesis. Here, we examined the role of cilostazol in the regulation of SREBP-1c transcription in the liver. The effects of cilostazol on the expression of SREBP-1c and its target genes in response to insulin or an LXR agonist (T0901317) were examined using real-time RT-PCR and western blot analysis on cultured hepatocytes. To investigate the effect of cilostazol on SREBP-1c at the transcriptional level, transient transfection reporter assays and electrophoretic mobility shift assays (EMSAs) were performed. Cilostazol inhibited insulin-induced and LXR-agonist-induced expression of SREBP-1c and its downstream targets, acetyl-CoA carboxylase and fatty acid synthase, in cultured hepatocytes. Cilostazol also inhibited activation of the SREBP-1c promoter by insulin, T0901317 and Sp1 in a luciferase reporter assay. EMSA analysis showed that cilostazol inhibits SREBP-1c expression by repressing the binding of LXR and Sp1 to the promoter region. These results indicate that cilostazol inhibits insulin-induced hepatic SREBP-1c expression via the inhibition of LXR and Sp1 activity and that cilostazol is a negative regulator of hepatic lipogenesis.
Collapse
|
49
|
Yang M, Liu W, Pellicane C, Sahyoun C, Joseph BK, Gallo-Ebert C, Donigan M, Pandya D, Giordano C, Bata A, Nickels JT. Identification of miR-185 as a regulator of de novo cholesterol biosynthesis and low density lipoprotein uptake. J Lipid Res 2013; 55:226-38. [PMID: 24296663 DOI: 10.1194/jlr.m041335] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Dysregulation of cholesterol homeostasis is associated with various metabolic diseases, including atherosclerosis and type 2 diabetes. The sterol response element binding protein (SREBP)-2 transcription factor induces the expression of genes involved in de novo cholesterol biosynthesis and low density lipoprotein (LDL) uptake, thus it plays a crucial role in maintaining cholesterol homeostasis. Here, we found that overexpressing microRNA (miR)-185 in HepG2 cells repressed SREBP-2 expression and protein level. miR-185-directed inhibition caused decreased SREBP-2-dependent gene expression, LDL uptake, and HMG-CoA reductase activity. In addition, we found that miR-185 expression was tightly regulated by SREBP-1c, through its binding to a single sterol response element in the miR-185 promoter. Moreover, we found that miR-185 expression levels were elevated in mice fed a high-fat diet, and this increase correlated with an increase in total cholesterol level and a decrease in SREBP-2 expression and protein. Finally, we found that individuals with high cholesterol had a 5-fold increase in serum miR-185 expression compared with control individuals. Thus, miR-185 controls cholesterol homeostasis through regulating SREBP-2 expression and activity. In turn, SREBP-1c regulates miR-185 expression through a complex cholesterol-responsive feedback loop. Thus, a novel axis regulating cholesterol homeostasis exists that exploits miR-185-dependent regulation of SREBP-2 and requires SREBP-1c for function.
Collapse
Affiliation(s)
- Muhua Yang
- The Institute of Metabolic Disorders and Genesis Biotechnology Group, Hamilton, NJ 08691
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Boden G, Salehi S, Cheung P, Homko C, Song W, Loveland-Jones C, Jayarajan S. Comparison of in vivo effects of insulin on SREBP-1c activation and INSIG-1/2 in rat liver and human and rat adipose tissue. Obesity (Silver Spring) 2013; 21:1208-14. [PMID: 23913732 PMCID: PMC3740458 DOI: 10.1002/oby.20134] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE The stimulatory effects of insulin on de novo lipogenesis (DNL) in the liver, where it is an important contributor to non-alcoholic fatty liver disease (NAFLD), hepatic and systemic insulin resistance, is strong and well established. In contrast, insulin plays only a minor role in DNL in adipose tissue. The reason why insulin stimulates DNL more in liver than in fat is not known but may be due to differential regulation of the transcription and post-translational activation of sterol regulatory element binding proteins (SREBPs). To test this hypothesis, we have examined effects of insulin on activation of SREBP-1c in liver of rats and in adipose tissue of rats and human subjects. DESIGN AND METHODS Liver and epidydimal fat were obtained from alert rats and subcutaneous adipose tissue from human subjects in response to 4 h euglycemic-hyperinsulinemic clamps. RESULTS Here we show that acutely raising plasma insulin levels in rats and humans increased SREBP-1 mRNA comparably 3-4 fold in rat liver and rat and human adipose tissue, but increased post-translational activation of SREBP-1c only in rat liver, while decreasing it in adipose tissue. These differential effects of insulin on SREBP-1c activation in liver and adipose tissue were associated with robust changes in the opposite direction of INSIG-1 and to a lesser extent of INSIG-2 mRNA and proteins. CONCLUSIONS We conclude that these findings support the hypothesis that insulin stimulated activation of SREBP-1c in the liver, at least in part, by suppressing INSIG-1 and -2, whereas in adipose tissue, an increase in INSIG-1 and -2 prevented SREBP-1c activation.
Collapse
Affiliation(s)
- Guenther Boden
- Division of Endocrinology/Diabetes/Metabolism, Clinical Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, USA.
| | | | | | | | | | | | | |
Collapse
|