1
|
Collins J, Piscopio RA, Reyland ME, Johansen CG, Benninger RKP, Farnsworth NL. Cleavage of protein kinase c δ by caspase-3 mediates proinflammatory cytokine-induced apoptosis in pancreatic islets. J Biol Chem 2024; 300:107611. [PMID: 39074637 PMCID: PMC11381875 DOI: 10.1016/j.jbc.2024.107611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 07/04/2024] [Accepted: 07/17/2024] [Indexed: 07/31/2024] Open
Abstract
In type 1 diabetes (T1D), autoreactive immune cells infiltrate the pancreas and secrete proinflammatory cytokines that initiate cell death in insulin producing islet β-cells. Protein kinase C δ (PKCδ) plays a role in mediating cytokine-induced β-cell death; however, the exact mechanisms are not well understood. To address this, we used an inducible β-cell specific PKCδ KO mouse as well as a small peptide inhibitor of PKCδ. We identified a role for PKCδ in mediating cytokine-induced β-cell death and have shown that inhibiting PKCδ protects pancreatic β-cells from cytokine-induced apoptosis in both mouse and human islets. We determined that cytokines induced nuclear translocation and activity of PKCδ and that caspase-3 cleavage of PKCδ may be required for cytokine-mediated islet apoptosis. Further, cytokine activated PKCδ increases activity both of proapoptotic Bax with acute treatment and C-Jun N-terminal kinase with prolonged treatment. Overall, our results suggest that PKCδ mediates cytokine-induced apoptosis via nuclear translocation, cleavage by caspase-3, and upregulation of proapoptotic signaling in pancreatic β-cells. Combined with the protective effects of PKCδ inhibition with δV1-1, the results of this study will aid in the development of novel therapies to prevent or delay β-cell death and preserve β-cell function in T1D.
Collapse
Affiliation(s)
- Jillian Collins
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, Colorado, USA
| | - Robert A Piscopio
- Barbara Davis Center for Childhood Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Mary E Reyland
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Chelsea G Johansen
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, Colorado, USA
| | - Richard K P Benninger
- Barbara Davis Center for Childhood Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.
| | - Nikki L Farnsworth
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, Colorado, USA; Barbara Davis Center for Childhood Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.
| |
Collapse
|
2
|
Ghoshal K, Luther JM, Pakala SB, Chetyrkin S, Falck JR, Zent R, Wasserman DH, Pozzi A. Epoxygenase Cyp2c44 Regulates Hepatic Lipid Metabolism and Insulin Signaling by Controlling FATP2 Localization and Activation of the DAG/PKCδ Axis. Diabetes 2024; 73:1229-1243. [PMID: 38743615 PMCID: PMC11262046 DOI: 10.2337/db23-0493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 05/05/2024] [Indexed: 05/16/2024]
Abstract
Cytochrome P450 epoxygenase Cyp2c44, a murine epoxyeicosatrienoic acid (EET)-producing enzyme, promotes insulin sensitivity, and Cyp2c44-/- mice show hepatic insulin resistance. Because insulin resistance leads to hepatic lipid accumulation and hyperlipidemia, we hypothesized that Cyp2c44 regulates hepatic lipid metabolism. Standard chow diet (SCD)-fed male Cyp2c44-/- mice had significantly decreased EET levels and increased hepatic and plasma lipid levels compared with wild-type mice. We showed increased hepatic plasma membrane localization of the FA transporter 2 (FATP2) and total unsaturated fatty acids and diacylglycerol (DAG) levels. Cyp2c44-/- mice had impaired glucose tolerance and increased hepatic plasma membrane-associated PKCδ and phosphorylated IRS-1, two negative regulators of insulin signaling. Surprisingly, SCD and high-fat diet (HFD)-fed Cyp2c44-/- mice had similar glucose tolerance and hepatic plasma membrane PKCδ levels, suggesting that SCD-fed Cyp2c44-/- mice have reached their maximal glucose intolerance. Inhibition of PKCδ resulted in decreased IRS-1 serine phosphorylation and improved insulin-mediated signaling in Cyp2c44-/- hepatocytes. Finally, Cyp2c44-/- HFD-fed mice treated with the analog EET-A showed decreased hepatic plasma membrane FATP2 and PCKδ levels with improved glucose tolerance and insulin signaling. In conclusion, loss of Cyp2c44 with concomitant decreased EET levels leads to increased hepatic FATP2 plasma membrane localization, DAG accumulation, and PKCδ-mediated attenuation of insulin signaling. Thus, Cyp2c44 acts as a regulator of lipid metabolism by linking it to insulin signaling. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Kakali Ghoshal
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - James M Luther
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - Suman B Pakala
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - Sergei Chetyrkin
- Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, TN
| | | | - Roy Zent
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
- Department of Veterans Affairs, Nashville, Nashville, TN
| | - David H Wasserman
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| | - Ambra Pozzi
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
- Department of Veterans Affairs, Nashville, Nashville, TN
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| |
Collapse
|
3
|
Zhong H, Sian V, Johari M, Katayama S, Oghabian A, Jonson PH, Hackman P, Savarese M, Udd B. Revealing myopathy spectrum: integrating transcriptional and clinical features of human skeletal muscles with varying health conditions. Commun Biol 2024; 7:438. [PMID: 38600180 PMCID: PMC11006663 DOI: 10.1038/s42003-024-06143-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 04/03/2024] [Indexed: 04/12/2024] Open
Abstract
Myopathy refers to a large group of heterogeneous, rare muscle diseases. Bulk RNA-sequencing has been utilized for the diagnosis and research of these diseases for many years. However, the existing valuable sequencing data often lack integration and clinical interpretation. In this study, we integrated bulk RNA-sequencing data from 1221 human skeletal muscles (292 with myopathies, 929 controls) from both databases and our local samples. By applying a method similar to single-cell analysis, we revealed a general spectrum of muscle diseases, ranging from healthy to mild disease, moderate muscle wasting, and severe muscle disease. This spectrum was further partly validated in three specific myopathies (97 muscles) through clinical features including trinucleotide repeat expansion, magnetic resonance imaging fat fraction, pathology, and clinical severity scores. This spectrum helped us identify 234 genuinely healthy muscles as unprecedented controls, providing a new perspective for deciphering the hallmark genes and pathways among different myopathies. The newly identified featured genes of general myopathy, inclusion body myositis, and titinopathy were highly expressed in our local muscles, as validated by quantitative polymerase chain reaction.
Collapse
Affiliation(s)
- Huahua Zhong
- Department of Neurology, Huashan Rare Disease Center, Huashan Hospital, Fudan University, Shanghai, China.
| | - Veronica Sian
- Department of Precision Medicine, "Luigi Vanvitelli" University of Campania, Via L. De Crecchio 7, Naples, Italy
| | - Mridul Johari
- Department of Medical and Clinical Genetics, Folkhälsan Research Center, Medicum, University of Helsinki, Helsinki, Finland
- Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands, WA, Australia
| | - Shintaro Katayama
- Department of Medical and Clinical Genetics, Folkhälsan Research Center, Medicum, University of Helsinki, Helsinki, Finland
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Ali Oghabian
- Department of Medical and Clinical Genetics, Folkhälsan Research Center, Medicum, University of Helsinki, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Per Harald Jonson
- Department of Medical and Clinical Genetics, Folkhälsan Research Center, Medicum, University of Helsinki, Helsinki, Finland
| | - Peter Hackman
- Department of Medical and Clinical Genetics, Folkhälsan Research Center, Medicum, University of Helsinki, Helsinki, Finland
| | - Marco Savarese
- Department of Medical and Clinical Genetics, Folkhälsan Research Center, Medicum, University of Helsinki, Helsinki, Finland
| | - Bjarne Udd
- Department of Medical and Clinical Genetics, Folkhälsan Research Center, Medicum, University of Helsinki, Helsinki, Finland
- Tampere Neuromuscular Center, University Hospital, Tampere, Finland
| |
Collapse
|
4
|
Kim Y, Kim HK, Kang S, Kim H, Go GW. Rottlerin suppresses lipid accumulation by inhibiting de novo lipogenesis and adipogenesis via LRP6/mTOR/SREBP1C in 3T3-L1 adipocytes. Food Sci Biotechnol 2023; 32:1445-1452. [PMID: 37457404 PMCID: PMC10349001 DOI: 10.1007/s10068-023-01339-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/24/2023] [Accepted: 05/12/2023] [Indexed: 07/18/2023] Open
Abstract
Rottlerin is isolated from Mallotus japonicus, a plant rich in polyphenols. Rottlerin is a selective PKCδ-inhibitor and is also known as an uncoupler of oxidative phosphorylation and anti-neoplastic agent. However, its anti-obesity effect is yet to be established. Therefore, this study tested whether rottlerin inhibits adipogenesis and de novo lipogenesis via the LRP6/mTOR/SREBP1C pathway in 3T3-L1 adipocytes. Rottlerin dramatically decreased lipid accumulation assessed by Oil Red O as evidence to support the cellular phenotype (p < 0.001). Pivotal messenger RNA and protein expressions associated with de novo lipogenesis (SREBP1C, ACC1, FAS, and SCD1) and adipogenesis (PPARγ and C/EBPα) were subsequentially verified by rottlerin in a dose-dependent manner (p < 0.05). Further investigation revealed that rottlerin reduced the AKT/mTOR pathway via diminished total protein of LRP6 (p < 0.05). Collectively, these findings establish a causal link between rottlerin, LRP6, and the altered nutrient-sensing mTOR pathway, in which rottlerin regulates de novo lipogenesis and adipogenesis in white adipocytes.
Collapse
Affiliation(s)
- Yejin Kim
- Department of Food and Nutrition, Hanyang University, Seoul, 04763 Republic of Korea
| | - Hyun Kyung Kim
- Department of Food and Nutrition, Hanyang University, Seoul, 04763 Republic of Korea
| | - Sumin Kang
- Department of Food and Nutrition, Hanyang University, Seoul, 04763 Republic of Korea
| | - Hayoon Kim
- Department of Food and Nutrition, Hanyang University, Seoul, 04763 Republic of Korea
| | - Gwang-woong Go
- Department of Food and Nutrition, Hanyang University, Seoul, 04763 Republic of Korea
| |
Collapse
|
5
|
LPIN1 Induces Gefitinib Resistance in EGFR Inhibitor-Resistant Non-Small Cell Lung Cancer Cells. Cancers (Basel) 2022; 14:cancers14092222. [PMID: 35565351 PMCID: PMC9102170 DOI: 10.3390/cancers14092222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/24/2022] [Accepted: 04/27/2022] [Indexed: 12/04/2022] Open
Abstract
Drug resistance limits the efficacy of targeted therapies, including tyrosine kinase inhibitors (TKIs); however, a substantial portion of the drug resistance mechanisms remains unexplained. In this study, we identified LPIN1 as a key factor that regulates gefitinib resistance in epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer (NSCLC) cells. Unlike TKI-sensitive HCC827 cells, gefitinib treatment induced LPIN1 expression and increased diacylglycerol concentration in TKI-resistant H1650 cells, followed by the activation of protein kinase C delta and nuclear factor kappa B (NF-κB) in an LPIN1-dependent manner, resulting in cancer cell survival. Additionally, LPIN1 increased the production of lipid droplets, which play an important role in TKI drug resistance. All results were recapitulated in a patient-derived EGFR-mutant NSCLC cell line. In in vivo tumorigenesis assay, we identified that both shRNA-mediated depletion and pharmaceutical inhibition of LPIN1 clearly reduced tumor growth and confirmed that gefitinib treatment induced LPIN1 expression and LPIN1-dependent NF-κB activation (an increase in p-IκBα level) in tumor tissues. These results suggest an effective strategy of co-treating TKIs and LPIN1 inhibitors to prevent TKI resistance in NSCLC patients.
Collapse
|
6
|
Recombinant humanized IgG1 maintain liver triglyceride homeostasis through Arylacetamide deacetylase in ApoE -/- mice. Int Immunopharmacol 2022; 108:108741. [PMID: 35397394 DOI: 10.1016/j.intimp.2022.108741] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 03/11/2022] [Accepted: 03/28/2022] [Indexed: 11/21/2022]
Abstract
BACKGROUND & AIMS Hyperlipidemia is a lipid metabolism disorder associated with elevated serum triglyceride (TG) and/or cholesterol. Over the years, studies have shown that hyperlipidemia is associated with combordities, incluing diabetes and obesity, gradually becoming a public health concern. Current treatment approaches remain limited due to the lack of effective drugs. Here we investigated the function of recombinant humanized IgG1 in maintaining liver TG homeostasis and the underlying mechanisms. METHODS ApoE-/- mice were fed a high-fat diet (HFD) for 20 weeks to induce hyperlipidemia. RNA sequencing (RNA-Seq) was performed to identify differences in gene expression in different groups of ApoE-/- mice liver. In vitro lipid accumulation in primary mouse hepatocytes was induced using a free fatty acid (FFA) mixture. Gene and protein expression were assessed in primary mouse hepatocytes by qPCR and Western blot. Gene reporter assays and ChIP-PCR were used to determine arylacetamide deacetylase (Aadac) promoter activity. RESULTS Recombinant humanized IgG1 could significantly decrease the serum level of TG and low-density lipoproteins (LDL-C). Moreover, hepatic TG and lipid droplets were also reduced compared to the HFD group. Mouse liver RNA-Seq revealed that administration of recombinant humanized IgG1 significantly elevated the expression of Aadac. In vitro, knock-down of Aadac could nullify the effect of recombinant humanized IgG1 on decreasing the lipid droplets induced by FFA in primary mouse hepatocytes. Gene Reporter assays and ChIP-PCR demonstrated that the foxa1 response element in the Aadac promoter played a key role in Aadac expression induced by recombinant humanized IgG1. Moreover, recombinant humanized IgG1 repressed phosphorylation of PKCδ and resulted in foxa1 elevation. Finally, neonatal Fc receptor (FcRn) knock-down reversed the effect of recombinant humanized IgG1 on the expression of PKCδ phosphorylation, foxa1 and Aadac. CONCLUSIONS Our findings suggest that recombinant humanized IgG1 plays an important role in maintaining liver TG homeostasis via the FcRn/PKCδ/foxa1/Aadac pathway.
Collapse
|
7
|
Mahmoudi A, Butler AE, Majeed M, Banach M, Sahebkar A. Investigation of the Effect of Curcumin on Protein Targets in NAFLD Using Bioinformatic Analysis. Nutrients 2022; 14:nu14071331. [PMID: 35405942 PMCID: PMC9002953 DOI: 10.3390/nu14071331] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/15/2022] [Accepted: 03/21/2022] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND: Non-alcoholic fatty liver disease (NAFLD) is a prevalent metabolic disorder. Defects in function/expression of genes/proteins are critical in initiation/progression of NAFLD. Natural products may modulate these genes/proteins. Curcumin improves steatosis, inflammation, and fibrosis progression. Here, bioinformatic tools, gene−drug and gene-disease databases were utilized to explore targets, interactions, and pathways through which curcumin could impact NAFLD. METHODS: Significant curcumin−protein interaction was identified (high-confidence:0.7) in the STITCH database. Identified proteins were investigated to determine association with NAFLD. gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were analyzed for significantly involved targets (p < 0.01). Specificity of obtained targets with NAFLD was estimated and investigated in Tissue/Cells−gene associations (PanglaoDB Augmented 2021, Mouse Gene Atlas) and Disease−gene association-based EnrichR algorithms (Jensen DISEASES, DisGeNET). RESULTS: Two collections were constructed: 227 protein−curcumin interactions and 95 NAFLD-associated genes. By Venn diagram, 14 significant targets were identified, and their biological pathways evaluated. Based on gene ontology, most targets involved stress and lipid metabolism. KEGG revealed chemical carcinogenesis, the AGE-RAGE signaling pathway in diabetic complications and NAFLD as the most common significant pathways. Specificity to diseases database (EnrichR algorithm) revealed specificity for steatosis/steatohepatitis. CONCLUSION: Curcumin may improve, or inhibit, progression of NAFLD through activation/inhibition of NAFLD-related genes.
Collapse
Affiliation(s)
- Ali Mahmoudi
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad 9177899191, Iran;
| | - Alexandra E. Butler
- Research Department, Royal College of Surgeons in Ireland Bahrain, Adliya 15503, Bahrain;
| | | | - Maciej Banach
- Nephrology and Hypertension, Department of Preventive Cardiology and Lipidology, Medical University of Lodz, 93-338 Lodz, Poland
- Cardiovascular Research Centre, University of Zielona Gora, 65-417 Zielona Gora, Poland
- Correspondence: (M.B.); (A.S.)
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9177899191, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad 9177899191, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177899191, Iran
- Correspondence: (M.B.); (A.S.)
| |
Collapse
|
8
|
Yan S, Sun M, Gao L, Yao N, Feng T, Yang Y, Li X, Hu W, Cui W, Li B. Identification of Key LncRNAs and Pathways in Prediabetes and Type 2 Diabetes Mellitus for Hypertriglyceridemia Patients Based on Weighted Gene Co-Expression Network Analysis. Front Endocrinol (Lausanne) 2022; 12:800123. [PMID: 35140684 PMCID: PMC8818867 DOI: 10.3389/fendo.2021.800123] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/13/2021] [Indexed: 12/19/2022] Open
Abstract
Aims Prevalence of prediabetes and type 2 diabetes mellitus(T2DM) are increasing worldwide. Key lncRNAs were detected to provide a reference for searching potential biomarkers of prediabetes and T2DM in hypertriglyceridemia patients. Methods The study included 18 hypertriglyceridemia patients: 6 newly diagnosed type 2 diabetes patients, 6 samples with prediabetes and 6 samples with normal blood glucose. Weighted gene co-expression network analysis (WGCNA) was conducted to construct co-expression network and obtain modules related to blood glucose, thus detecting key lncRNAs. Results The green, yellow and yellow module was significantly related to blood glucose in T2DM versus normal controls, T2DM versus prediabetes, prediabetes versus normal controls, respectively. ENST00000503273, ENST00000462720, ENST00000480633 and ENST00000485392 were detected as key lncRNAs for the above three groups, respectively. Conclusions For hypertriglyceridemia patients with different blood glucose levels, ENST00000503273, ENST00000462720 and ENST00000480633 could be potential biomarkers of T2DM.
Collapse
Affiliation(s)
- Shoumeng Yan
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, China
| | - Mengzi Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, China
| | - Lichao Gao
- Department of Endocrinology, The First Hospital of Jilin University, Changchun, China
| | - Nan Yao
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, China
| | - Tianyu Feng
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, China
| | - Yixue Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, China
| | - Xiaotong Li
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, China
| | - Wenyu Hu
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, China
| | - Weiwei Cui
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, China
| | - Bo Li
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, China
| |
Collapse
|
9
|
Small L, Ehrlich A, Iversen J, Ashcroft SP, Trošt K, Moritz T, Hartmann B, Holst JJ, Treebak JT, Zierath JR, Barrès R. Comparative analysis of oral and intraperitoneal glucose tolerance tests in mice. Mol Metab 2022; 57:101440. [PMID: 35026435 PMCID: PMC8810558 DOI: 10.1016/j.molmet.2022.101440] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 01/06/2022] [Accepted: 01/08/2022] [Indexed: 01/18/2023] Open
Abstract
Objective The glucose tolerance test (GTT) is widely used in preclinical research to investigate glucose metabolism, but there is no standardised way to administer glucose. The aim of this study was to directly compare the effect of the route of glucose administration on glucose and insulin kinetics during a GTT in mice. Methods A GTT was performed in lean male and female mice and obese male mice and glucose was administered via the oral or intraperitoneal (I.P.) route. Samples were collected frequently during the GTT to provide a full time-course of the insulin and glucose excursions. In another cohort of lean male mice, plasma concentrations of insulin, c-peptide, and incretin hormones were measured at early time points after glucose administration. A stable-isotope labelled GTT (SiGTT) was then performed to delineate the contribution of exogenous and endogenous glucose to glycemia during the GTT, comparing both methods of glucose administration. Finally, we present a method to easily measure insulin from small volumes of blood during a GTT by directly assaying whole-blood insulin using ELISA and show a good concordance between whole-blood and plasma insulin measurements. Results We report that I.P. glucose administration results in an elevated blood glucose excursion and a largely absent elevation in blood insulin and plasma incretin hormones when compared to oral administration. Utilising stable-isotope labelled glucose, we demonstrate that the difference in glucose excursion between the two routes of administration is mainly due to the lack of suppression of glucose production in I.P. injected mice. Additionally, rates of exogenous glucose appearance into circulation were different between lean and obese mice after I.P., but not after oral glucose administration. Conclusion Reflecting on these data, we suggest that careful consideration be given to the route of glucose administration when planning a GTT procedure in mice and that in most circumstances the oral route of glucose administration should be preferred over the I.P. route to avoid possible artifacts originating from a non-physiological route. Intraperitoneal glucose administration does not promote insulin secretion. Exogenous glucose appearance is delayed in obese mice after intraperitoneal administration. Hepatic glucose production is suppressed after administering oral not intraperitoneal glucose. Measuring insulin from whole blood is comparable to that from plasma.
Collapse
Affiliation(s)
- Lewin Small
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen
| | - Amy Ehrlich
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen
| | - Jo Iversen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen
| | - Stephen P Ashcroft
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen
| | - Kajetan Trošt
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen
| | - Thomas Moritz
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen; Swedish Metabolomics Centre, Department of Plant Physiology and Forest Genetics, Swedish University of Agricultural Sciences
| | - Bolette Hartmann
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen; Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen
| | - Jens J Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen; Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen
| | - Juleen R Zierath
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen; Department of Physiology and Pharmacology and Section for Integrative Physiology, Department of Molecular Medicine and Surgery and Karolinska Institutet
| | - Romain Barrès
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen; Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d'Azur and CNRS.
| |
Collapse
|
10
|
Yin H, Shi A, Wu J. Platelet-Activating Factor Promotes the Development of Non-Alcoholic Fatty Liver Disease. Diabetes Metab Syndr Obes 2022; 15:2003-2030. [PMID: 35837578 PMCID: PMC9275506 DOI: 10.2147/dmso.s367483] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/28/2022] [Indexed: 11/23/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a multifaceted clinicopathological syndrome characterised by excessive hepatic lipid accumulation that causes steatosis, excluding alcoholic factors. Platelet-activating factor (PAF), a biologically active lipid transmitter, induces platelet activation upon binding to the PAF receptor. Recent studies have found that PAF is associated with gamma-glutamyl transferase, which is an indicator of liver disease. Moreover, PAF can stimulate hepatic lipid synthesis and cause hypertriglyceridaemia. Furthermore, the knockdown of the PAF receptor gene in the animal models of NAFLD helped reduce the inflammatory response, improve glucose homeostasis and delay the development of NAFLD. These findings suggest that PAF is associated with NAFLD development. According to reports, patients with NAFLD or animal models have marked platelet activation abnormalities, mainly manifested as enhanced platelet adhesion and aggregation and altered blood rheology. Pharmacological interventions were accompanied by remission of abnormal platelet activation and significant improvement in liver function and lipids in the animal model of NAFLD. These confirm that platelet activation may accompany a critical importance in NAFLD development and progression. However, how PAFs are involved in the NAFLD signalling pathway needs further investigation. In this paper, we review the relevant literature in recent years and discuss the role played by PAF in NAFLD development. It is important to elucidate the pathogenesis of NAFLD and to find effective interventions for treatment.
Collapse
Affiliation(s)
- Hang Yin
- Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, People’s Republic of China
| | - Anhua Shi
- Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, People’s Republic of China
| | - Junzi Wu
- Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, People’s Republic of China
- Correspondence: Junzi Wu; Anhua Shi, Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, People’s Republic of China, Tel/Fax +86 187 8855 7524; +86 138 8885 0813, Email ;
| |
Collapse
|
11
|
Schmitz-Peiffer C. Deconstructing the Role of PKC Epsilon in Glucose Homeostasis. Trends Endocrinol Metab 2020; 31:344-356. [PMID: 32305097 DOI: 10.1016/j.tem.2020.01.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 01/22/2020] [Accepted: 01/24/2020] [Indexed: 02/06/2023]
Abstract
The failure of insulin to suppress glucose production by the liver is a key aspect of the insulin resistance seen in type 2 diabetes. Lipid-activated protein kinase C epsilon has long been identified as an important mediator of diet-induced glucose intolerance and hepatic insulin resistance and the current view emphasizes a mechanism involving phosphorylation of the insulin receptor by the kinase to inhibit downstream insulin action. However, the significance of this direct effect in the liver has now been challenged by tissue-specific deletion of PKCε, which demonstrated a more prominent role for the kinase in adipose tissue to promote glucose intolerance. New insights regarding the role of PKCε therefore contribute to the understanding of indirect effects on hepatic glucose metabolism.
Collapse
Affiliation(s)
- Carsten Schmitz-Peiffer
- Garvan Institute of Medical Research, Darlinghurst Sydney, NSW 2010, Australia; St Vincent's Clinical School, University of New South Wales, Sydney, NSW 2010, Australia.
| |
Collapse
|
12
|
The role of proprotein convertase subtilisin-kexin type 9 (PCSK9) in the vascular aging process - is there a link? POLISH JOURNAL OF THORACIC AND CARDIOVASCULAR SURGERY 2019; 16:128-132. [PMID: 31708986 PMCID: PMC6836637 DOI: 10.5114/kitp.2019.88602] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 09/04/2019] [Indexed: 12/11/2022]
Abstract
Lately there are many new, promising low-density lipoprotein cholesterol reducing therapies with PCSK9 inhibitors. We performed selected sampling of the publications in PubMed and made a review according to selected keywords. It summarizes the effect of PCSK9 on vascular aging, directly associated with lipid and glucose metabolism, chronic inflammation, atherosclerosis and hypertension. Serum level of PCSK9 is different in patients affected by certain illnesses (whose risk increases with age) than in healthy individuals. The same could be observed in the case of chronic inflammation. In this review we summarize what is known about the role PCSK9 in human metabolism and how this could affect the vascular aging process. Based on the available sources, we prove that PCSK9 is involved in many biochemical pathways associated with vascular aging. In the future, treatments using PCSK9 inhibition may not only reduce the cardiovascular risk but also slow down this process.
Collapse
|
13
|
Rivers SL, Klip A, Giacca A. NOD1: An Interface Between Innate Immunity and Insulin Resistance. Endocrinology 2019; 160:1021-1030. [PMID: 30807635 PMCID: PMC6477778 DOI: 10.1210/en.2018-01061] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 02/19/2019] [Indexed: 12/17/2022]
Abstract
Insulin resistance is driven, in part, by activation of the innate immune system. We have discussed the evidence linking nucleotide-binding oligomerization domain (NOD)1, an intracellular pattern recognition receptor, to the onset and progression of obesity-induced insulin resistance. On a molecular level, crosstalk between downstream NOD1 effectors and the insulin receptor pathway inhibits insulin signaling, potentially through reduced insulin receptor substrate action. In vivo studies have demonstrated that NOD1 activation induces peripheral, hepatic, and whole-body insulin resistance. Also, NOD1-deficient models are protected from high-fat diet (HFD)-induced insulin resistance. Moreover, hematopoietic NOD1 deficiency prevented HFD-induced changes in proinflammatory macrophage polarization status, thus protecting against the development of metabolic inflammation and insulin resistance. Serum from HFD-fed mice activated NOD1 signaling ex vivo; however, the molecular identity of the activating factors remains unclear. Many have proposed that an HFD changes the gut permeability, resulting in increased translocation of bacterial fragments and increased circulating NOD1 ligands. In contrast, others have suggested that NOD1 ligands are endogenous and potentially lipid-derived metabolites produced during states of nutrient overload. Nevertheless, that NOD1 contributes to the development of insulin resistance, and that NOD1-based therapy might provide benefit, is an exciting advancement in metabolic research.
Collapse
Affiliation(s)
- Sydney L Rivers
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Amira Klip
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Adria Giacca
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Banting and Best Diabetes Centre, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Correspondence: Adria Giacca, MD, Department of Physiology, Faculty of Medicine, University of Toronto, Medical Sciences Building, 1 King’s College Circle, No. 3336, Toronto, Ontario M5S 1A8, Canada. E-mail:
| |
Collapse
|
14
|
Mulcahy MJ, Paulo JA, Hawrot E. Proteomic Investigation of Murine Neuronal α7-Nicotinic Acetylcholine Receptor Interacting Proteins. J Proteome Res 2018; 17:3959-3975. [PMID: 30285449 DOI: 10.1021/acs.jproteome.8b00618] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The α7-nicotinic acetylcholine receptor (α7-nAChR) is a ligand-gated ion channel that is expressed widely in vertebrates and is the principal high-affinity α-bungarotoxin (α-bgtx) binding protein in the mammalian CNS. α7-nAChRs associate with proteins that can modulate its properties. The α7-nAChR interactome is the summation of proteins interacting or associating with α7-nAChRs in a protein complex. To identify an α7-nAChR interactome in neural tissue, we isolated α-bgtx-affinity protein complexes from wild-type and α7-nAChR knockout (α7 KO) mouse whole brain tissue homogenates using α-bgtx-affinity beads. Affinity precipitated proteins were trypsinized and analyzed with an Orbitrap Fusion mass spectrometer. Proteins isolated with the α7-nAChR specific ligand, α-bgtx, were determined to be α7-nAChR associated proteins. The α7-nAChR subunit and 120 additional proteins were identified. Additionally, 369 proteins were identified as binding to α-bgtx in the absence of α7-nAChR expression, thereby identifying nonspecific proteins for α7-nAChR investigations using α-bgtx enrichment. These results expand on our previous investigations of α7-nAChR interacting proteins using α-bgtx-affinity bead isolation by controlling for differences between α7-nAChR and α-bgtx-specific proteins, developing an improved protein isolation methodology, and incorporating the latest technology in mass spectrometry. The α7-nAChR interactome identified in this study includes proteins associated with the expression, localization, function, or modulation of α7-nAChRs, and it provides a foundation for future studies to elucidate how these interactions contribute to human disease.
Collapse
Affiliation(s)
- Matthew J Mulcahy
- Division of Biology and Biological Engineering , California Institute of Technology , 1200 East California Boulevard , Pasadena , California 91125-2900 , United States.,Department of Molecular Pharmacology, Physiology and Biotechnology , Brown University , Providence , Rhode Island 02912 , United States
| | - Joao A Paulo
- Department of Cell Biology , Harvard Medical School , 240 Longwood Avenue , Boston , Massachusetts 02115 , United States
| | - Edward Hawrot
- Department of Molecular Pharmacology, Physiology and Biotechnology , Brown University , Providence , Rhode Island 02912 , United States
| |
Collapse
|
15
|
Sun S, Wu Q, Song J, Sun S. Protein kinase C δ-dependent regulation of Ubiquitin-proteasome system function in breast cancer. Cancer Biomark 2018; 21:1-9. [PMID: 29036789 DOI: 10.3233/cbm-170451] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Besides the crucial role of hyperinsulinemia in the development of breast cancer with Type 2 diabetes mellitus (T2DM), it has been shown that hyperglycemia could contribute to promote cancer progression. A remarkable association within hyperglycemia, PKCδ and Ubiquitin-proteasome system (UPS) has been reported, suggesting that PKCδ may mediate high glucose-induced UPS activation in breast cancer cells. Although the independent effects of PKCδ or UPS on breast cancer and T2DM are increasingly supported by experimental evidence, the complex interactional link between PKCδ and UPS is still unclear. Hence, we focus on the relationship between PKCδ and UPS in breast cancer with T2DM. We hypothesize that PKCδ may have the function to regulate the activity of UPS. Further, we speculate that PKCδ combine with proteasome α2 promoter, that indicate PKCδ regulate the function of UPS by change the composition of proteasome. Therefore, we surmise that PKCδ mediated high glucose-induced UPS activation in breast cancer cells, and specific PKCδ inhibitor rottlerin significantly suppressed elevated glucose induced the activity of UPS. We hope that our paper will stimulate further studies the relationship between PKCδ and UPS, and a new targeted therapy and early medical intervention for PKCδ could be a useful option for breast cancer cases complicated with T2DM or hyperglycemia.
Collapse
Affiliation(s)
- Si Sun
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qi Wu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Junlong Song
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Shengrong Sun
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
16
|
Dallak MA. Acylated ghrelin induces but deacylated ghrelin prevents hepatic steatosis and insulin resistance in lean rats: Effects on DAG/ PKC/JNK pathway. Biomed Pharmacother 2018; 105:299-311. [PMID: 29860222 DOI: 10.1016/j.biopha.2018.05.098] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 05/20/2018] [Accepted: 05/21/2018] [Indexed: 12/19/2022] Open
Abstract
This study investigated the molecular effects of acylated (AG) and unacylated ghrelin (UAG) or their combination on hepatic lipogenesis pathways and DAG/PKC/JNK signaling in the livers of lean rats fed standard diet. Male rats (n = 10) were classified as control + vehicle (saline, 200 μl), AG, UAG, and AG + UAG-treated groups. All treatments were given at final doses of 200 ng/kg of for 14 days (twice/day, S.C). Administration of AG significantly enhanced circulatory levels of AG and UAG turning the normal ratio of AG/UAG from 1:2.5 to 1:1.2. However, while UAG didn't affect circulatory levels of AG, administration of UAG alone or in combination with AG resulted in AG/UAG ratios of 1:7 and 1:3, respectively. Independent of food intake nor the development of peripheral IR, AG increased hepatic DAG, TGs and CHOL contents and induced hepatic IR. Mechanism of action include 1) upregulation of mRNA and protein levels of DGAT-2 and mtGPAT-1, SREBP-1 and SCD-1, and 2) inhibition of fatty acids (FAs) oxidation mediated by inhibition of AMPK/ PPAR-α/CPT-1 axis. Consequently, AG induced membranous translocation of PKCδ and PKCε leading to activation of JNK and significant inhibition of insulin signaling under basal and insulin stimulation as evident by decreases in the phosphorylation levels of IRS (Tyr612) and Akt (Thr318) and increased phosphorylation of IRS (Ser307). However, while UAG only activated FAs oxidation in control rats, it reversed all alterations in all measured biochemical endpoints seen in the AG-treated group, when administered in combination with AG, leading to significant decreases in hepatic fat accumulation and prevention of hepatic IR. In conclusion, while exogenous administration of AG is at high risk of developing steatohepatitis and hepatic IR, co-administration of a balanced dose of UAG reduces this risk and inhibits hepatic lipid accumulation and enhance hepatic insulin signaling.
Collapse
Affiliation(s)
- Mohammad A Dallak
- Department of Physiology, College of Medicine, King's Khalid University, Abha, 61241, Saudi Arabia.
| |
Collapse
|
17
|
Parihar SP, Ozturk M, Marakalala MJ, Loots DT, Hurdayal R, Maasdorp DB, Van Reenen M, Zak DE, Darboe F, Penn-Nicholson A, Hanekom WA, Leitges M, Scriba TJ, Guler R, Brombacher F. Protein kinase C-delta (PKCδ), a marker of inflammation and tuberculosis disease progression in humans, is important for optimal macrophage killing effector functions and survival in mice. Mucosal Immunol 2018; 11:496-511. [PMID: 28832027 DOI: 10.1038/mi.2017.68] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 06/13/2017] [Indexed: 02/04/2023]
Abstract
We previously demonstrated that protein kinase C-δ (PKCδ) is critical for immunity against Listeria monocytogenes, Leishmania major, and Candida albicans infection in mice. However, the functional relevance of PKCδ during Mycobacterium tuberculosis (Mtb) infection is unknown. PKCδ was significantly upregulated in whole blood of patients with active tuberculosis (TB) disease. Lung proteomics further revealed that PKCδ was highly abundant in the necrotic and cavitory regions of TB granulomas in multidrug-resistant human participants. In murine Mtb infection studies, PKCδ-/- mice were highly susceptible to tuberculosis with increased mortality, weight loss, exacerbated lung pathology, uncontrolled proinflammatory cytokine responses, and increased mycobacterial burdens. Moreover, these mice displayed a significant reduction in alveolar macrophages, dendritic cells, and decreased accumulation of lipid bodies (lungs and macrophages) and serum fatty acids. Furthermore, a peptide inhibitor of PKCδ in wild-type mice mirrored lung inflammation identical to infected PKCδ-/- mice. Mechanistically, increased bacterial growth in macrophages from PKCδ-/- mice was associated with a decline in killing effector functions independent of phagosome maturation and autophagy. Taken together, these data suggest that PKCδ is a marker of inflammation during active TB disease in humans and required for optimal macrophage killing effector functions and host protection during Mtb infection in mice.
Collapse
Affiliation(s)
- S P Parihar
- Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town-Component, Cape Town, South Africa
| | - M Ozturk
- Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town-Component, Cape Town, South Africa
| | - M J Marakalala
- Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - D T Loots
- Human Metabolomics, North-West University, Potchefstroom, South Africa
| | - R Hurdayal
- Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town-Component, Cape Town, South Africa.,Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, Cape Town, South Africa
| | - D Beukes Maasdorp
- Human Metabolomics, North-West University, Potchefstroom, South Africa
| | - M Van Reenen
- Human Metabolomics, North-West University, Potchefstroom, South Africa
| | - D E Zak
- Center for Infectious Disease Research, Seattle, WA, USA
| | - F Darboe
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine (IDM) & Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - A Penn-Nicholson
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine (IDM) & Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - W A Hanekom
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine (IDM) & Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - M Leitges
- PKC Research Consult, Cologne, Germany
| | - T J Scriba
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine (IDM) & Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - R Guler
- Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town-Component, Cape Town, South Africa
| | - F Brombacher
- Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town-Component, Cape Town, South Africa
| |
Collapse
|
18
|
Schmitz-Peiffer C. Anarchy in the UPR: A Ca 2+-insensitive PKC inhibits SERCA activity to promote ER stress. Biosci Rep 2018; 38:BSR20170966. [PMID: 29439143 PMCID: PMC5857902 DOI: 10.1042/bsr20170966] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 02/05/2018] [Accepted: 02/07/2018] [Indexed: 02/04/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is highly prevalent in Western countries, and is linked to the development of liver cancer and Type 2 diabetes (T2D). It is strongly associated with obesity, but the dysregulation of liver lipid storage is not fully understood. Fatty acid oversupply to hepatocytes can establish a vicious cycle involving diminished protein folding, endoplasmic reticulum (ER) stress, insulin resistance and further lipogenesis. This commentary discusses the recent findings of Lai et al. published in Bioscience Reports, that implicate protein kinase C delta (PKCδ) activation by fatty acids in the inhibition of the SERCA Ca2+ pump, resulting in reduced ER Ca2+ loading and protein misfolding. PKCδ therefore represents a target for the treatment of both steatosis and insulin resistance, key to the prevention of NAFLD and T2D.
Collapse
Affiliation(s)
- Carsten Schmitz-Peiffer
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, and St Vincents Clinical School, University of New South Wales, Darlinghurst, Sydney, 2010, Australia
| |
Collapse
|
19
|
de Castro GS, Calder PC. Non-alcoholic fatty liver disease and its treatment with n-3 polyunsaturated fatty acids. Clin Nutr 2018; 37:37-55. [DOI: 10.1016/j.clnu.2017.01.006] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 12/12/2016] [Accepted: 01/10/2017] [Indexed: 02/08/2023]
|
20
|
PKCδ silencing alleviates saturated fatty acid induced ER stress by enhancing SERCA activity. Biosci Rep 2017; 37:BSR20170869. [PMID: 29046367 PMCID: PMC5700272 DOI: 10.1042/bsr20170869] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 10/05/2017] [Accepted: 10/09/2017] [Indexed: 02/07/2023] Open
Abstract
Protein kinase C δ (PKCδ) plays an important role in nonalcoholic fatty liver disease (NAFLD), however, the mechanism remains unknown. The present study explored the role of PKCδ in NAFLD development and investigated the relationships between PKCδ, calcium homeostasis, and endoplasmic reticulum (ER) stress (ERS). Hepatic steatosis cell model was induced by palmitic acid (PA) in L02 cells. Lipid accretion was evaluated using Oil Red O staining and a triglyceride (TG) detection kit. PKCδ was down-regulated by siRNA. RT-PCR and Western blotting were used to detect the expression of ERS markers. The fluorescence of Ca2+ influx was recorded using confocal microscopy. Sarco-ER Ca2+-ATPase (SERCA) activity was measured by ultramicro-ATP enzyme test kit. PA treatment induced lipid accretion in L02 cells, destroyed the ER structure, and increased PKCδ activation in a time-dependent manner. Further, PA treatment significantly increased the expression of ERS markers, Ig heavy chain binding protein (Bip), and homologous proteins of CCAAT-enhancer binding proteins (CHOP). PKCδ silencing down-regulated Bip and CHOP expression, indicating a successful alleviation of ERS. The increased calcium storage induced by PA stimulation was significantly decreased in L02 cells treated with PKCδ siRNA compared with the negative control. Moreover, diminished SERCA activity caused by PA was recovered in PKCδ siRNA transfected cells. To the best of our knowledge, this is the first report demonstrating that the inhibition of PKCδ alleviates ERS by enhancing SERCA activity and stabilizing calcium homeostasis.
Collapse
|
21
|
Capuani B, Pacifici F, Pastore D, Palmirotta R, Donadel G, Arriga R, Bellia A, Di Daniele N, Rogliani P, Abete P, Sbraccia P, Guadagni F, Lauro D, Della-Morte D. The role of epsilon PKC in acute and chronic diseases: Possible pharmacological implications of its modulators. Pharmacol Res 2016; 111:659-667. [PMID: 27461137 DOI: 10.1016/j.phrs.2016.07.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 07/22/2016] [Indexed: 02/06/2023]
|
22
|
Liao BM, McManus SA, Hughes WE, Schmitz-Peiffer C. Flavin-Containing Monooxygenase 3 Reduces Endoplasmic Reticulum Stress in Lipid-Treated Hepatocytes. Mol Endocrinol 2016; 30:417-28. [PMID: 26886171 DOI: 10.1210/me.2015-1217] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Isoforms of flavin-containing monooxygenase (FMO) are involved in xenobiotic metabolism but have also been implicated in the regulation of glucose and lipid homeostasis and in the development of atherosclerosis. However, we have recently shown that improved insulin action is associated with increased FMO expression in livers of protein kinase C-deficient mice. Here, we investigated whether FMO3 expression affected insulin signaling, glucose metabolism, and endoplasmic reticulum (ER) stress in hepatocytes. HepG2 and IHH hepatocytes were transfected with FMO3 cDNA for overexpression, or small interfering RNA for knockdown. Cells were treated with palmitate to induce insulin resistance and insulin signaling, phosphoenolpyruvate carboxykinase (PEPCK) gene expression and ER stress markers were examined by immunoblotting and RT-PCR. Glycogen synthesis was measured using [(14)C]glucose. Palmitate treatment reduced insulin signaling at the level of Akt phosphorylation and glycogen synthesis, which were little affected by FMO3 overexpression. However, the fatty acid also increased the levels of several ER stress markers and activation of caspase 3, which were counteracted by FMO3 overexpression and exacerbated by FMO3 knockdown. Although FMO3 expression did not reverse lipid effects on protein thiol redox in hepatocytes, it did prevent up-regulation of the gluconeogenic enzyme PEPCK by pharmacological ER stress inducers or by palmitate. ER stress and PEPCK levels were also reduced in livers of fat-fed protein kinase Cδ-deficient mice. Our data indicate that FMO3 can contribute to the regulation of glucose metabolism in the liver by reducing lipid-induced ER stress and the expression of PEPCK, independently of insulin signal transduction.
Collapse
Affiliation(s)
- Bing M Liao
- Diabetes and Metabolism Division (B.M.L., S.A.M., W.E.H., C.S.-P.), Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia; and St. Vincent's Hospital Clinical School (W.E.H., C.S.-P.), Faculty of Medicine, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Sophie A McManus
- Diabetes and Metabolism Division (B.M.L., S.A.M., W.E.H., C.S.-P.), Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia; and St. Vincent's Hospital Clinical School (W.E.H., C.S.-P.), Faculty of Medicine, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - William E Hughes
- Diabetes and Metabolism Division (B.M.L., S.A.M., W.E.H., C.S.-P.), Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia; and St. Vincent's Hospital Clinical School (W.E.H., C.S.-P.), Faculty of Medicine, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Carsten Schmitz-Peiffer
- Diabetes and Metabolism Division (B.M.L., S.A.M., W.E.H., C.S.-P.), Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia; and St. Vincent's Hospital Clinical School (W.E.H., C.S.-P.), Faculty of Medicine, University of New South Wales, Sydney, New South Wales 2052, Australia
| |
Collapse
|
23
|
Seed Ahmed M, Ahmed MS, Pelletier J, Leumann H, Gu HF, Östenson CG. Expression of Protein Kinase C Isoforms in Pancreatic Islets and Liver of Male Goto-Kakizaki Rats, a Model of Type 2 Diabetes. PLoS One 2015; 10:e0135781. [PMID: 26398746 PMCID: PMC4580567 DOI: 10.1371/journal.pone.0135781] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 07/27/2015] [Indexed: 02/03/2023] Open
Abstract
Protein kinase C (PKC) is a family of protein kinases controlling protein phosphorylation and playing important roles in the regulation of metabolism. We have investigated expression levels of PKC isoforms in pancreatic islets and liver of diabetic Goto-Kakizaki (GK) rats with and without insulin treatment to evaluate their association with glucose homeostasis. mRNA and protein expression levels of PKC isoforms were assessed in pancreatic islets and liver of Wistar rats and GK rats with or without insulin treatment. PKCα and PKCζ mRNA expressions were down-regulated in islets of GK compared with Wistar rats. PKCα and phosphorylated PKCα (p-PKCα) protein expressions were decreased in islets of GK compared with insulin-treated GK and Wistar rats. PKCζ protein expression in islets was reduced in GK and insulin-treated GK compared with Wistar rats, but p-PKCζ was decreased only in GK rats. Islet PKCε mRNA and protein expressions were lower in GK compared with insulin-treated GK and Wistar rats. In liver, PKCδ and PKCζ mRNA expressions were decreased in both GK and insulin-treated GK compared with Wistar rats. Hepatic PKCζ protein expression was diminished in both GK rats with and without insulin treatment compared with Wistar rats. Hepatic PKCε mRNA expression was down-regulated in insulin-treated GK compared with GK and Wistar rats. PKCα, PKCε, and p-PKCζ expressions were secondary to hyperglycaemia in GK rat islets. Hepatic PKCδ and PKCζ mRNA expressions were primarily linked to hyperglycaemia. Additionally, hepatic PKCε mRNA expression could be under control of insulin.
Collapse
Affiliation(s)
- Mohammed Seed Ahmed
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, Solna, Stockholm, Sweden; Department of Physiology, Faculty of Medicine, University of Khartoum, Khartoum, Sudan
| | | | - Julien Pelletier
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Hannes Leumann
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Harvest F Gu
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Claes-Göran Östenson
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, Solna, Stockholm, Sweden
| |
Collapse
|
24
|
Does Diacylglycerol Accumulation in Fatty Liver Disease Cause Hepatic Insulin Resistance? BIOMED RESEARCH INTERNATIONAL 2015; 2015:104132. [PMID: 26273583 PMCID: PMC4529893 DOI: 10.1155/2015/104132] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 01/27/2015] [Indexed: 01/04/2023]
Abstract
Numerous studies conducted on obese humans and various rodent models of obesity have identified a correlation between hepatic lipid content and the development of insulin resistance in liver and other tissues. Despite a large body of the literature on this topic, the cause and effect relationship between hepatic steatosis and insulin resistance remains controversial. If, as many believe, lipid aggregation in liver drives insulin resistance and other metabolic abnormalities, there are significant unanswered questions as to which lipid mediators are causative in this cascade. Several published papers have now correlated levels of diacylglycerol (DAG), the penultimate intermediate in triglyceride synthesis, with development of insulin resistance and have postulated that this occurs via activation of protein kinase C signaling. Although many studies have confirmed this relationship, many others have reported a disconnect between DAG content and insulin resistance. It has been postulated that differences in methods for DAG measurement, DAG compartmentalization within the cell, or fatty acid composition of the DAG may explain these discrepancies. The purpose of this review is to compare and contrast some of the relevant findings in this area and to discuss a number of unanswered questions regarding the relationship between DAG and insulin resistance.
Collapse
|
25
|
Fabbrini E, Magkos F. Hepatic Steatosis as a Marker of Metabolic Dysfunction. Nutrients 2015; 7:4995-5019. [PMID: 26102213 PMCID: PMC4488828 DOI: 10.3390/nu7064995] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 06/05/2015] [Accepted: 06/15/2015] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the liver manifestation of the complex metabolic derangements associated with obesity. NAFLD is characterized by excessive deposition of fat in the liver (steatosis) and develops when hepatic fatty acid availability from plasma and de novo synthesis exceeds hepatic fatty acid disposal by oxidation and triglyceride export. Hepatic steatosis is therefore the biochemical result of an imbalance between complex pathways of lipid metabolism, and is associated with an array of adverse changes in glucose, fatty acid, and lipoprotein metabolism across all tissues of the body. Intrahepatic triglyceride (IHTG) content is therefore a very good marker (and in some cases may be the cause) of the presence and the degree of multiple-organ metabolic dysfunction. These metabolic abnormalities are likely responsible for many cardiometabolic risk factors associated with NAFLD, such as insulin resistance, type 2 diabetes mellitus, and dyslipidemia. Understanding the factors involved in the pathogenesis and pathophysiology of NAFLD will lead to a better understanding of the mechanisms responsible for the metabolic complications of obesity, and hopefully to the discovery of novel effective treatments for their reversal.
Collapse
Affiliation(s)
- Elisa Fabbrini
- Center for Human Nutrition and Atkins Center of Excellence in Obesity Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Faidon Magkos
- Center for Human Nutrition and Atkins Center of Excellence in Obesity Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
26
|
Berlanga A, Guiu-Jurado E, Porras JA, Aragonès G, Auguet T. [Role of metabolic lipases and lipotoxicity in the development of non-alcoholic steatosis and non-alcoholic steatohepatitis]. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS 2015; 28:47-61. [PMID: 26049666 DOI: 10.1016/j.arteri.2015.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 03/03/2015] [Accepted: 03/04/2015] [Indexed: 10/23/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become the most common liver disease in developed countries, covering a spectrum of pathological conditions ranging from single steatosis to non-alcoholic steatohepatitis, cirrhosis and hepatocellular carcinoma. Its pathogenesis has been often interpreted by the "double-hit" hypothesis, where the lipid accumulation in the liver is followed by proinflammatory mediators inducing inflammation, hepatocellular injury and fibrosis. Nowadays, a more complex model suggests that free fatty acids and their metabolites could be the true lipotoxic agents that contribute to the development of NAFLD and hepatic insulin resistance, suggesting a central role for metabolic lipases in that process.
Collapse
Affiliation(s)
- Alba Berlanga
- Grupo de recerca GEMMAIR (AGAUR)-Medicina Aplicada, Departamento de Medicina y Cirugía, Universidad Rovira i Virgili (URV), Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, España
| | - Esther Guiu-Jurado
- Grupo de recerca GEMMAIR (AGAUR)-Medicina Aplicada, Departamento de Medicina y Cirugía, Universidad Rovira i Virgili (URV), Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, España
| | - José Antonio Porras
- Grupo de recerca GEMMAIR (AGAUR)-Medicina Aplicada, Departamento de Medicina y Cirugía, Universidad Rovira i Virgili (URV), Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, España; Servicio de Medicina Interna, Hospital Universitario Joan XXIII, Tarragona, España
| | - Gemma Aragonès
- Grupo de recerca GEMMAIR (AGAUR)-Medicina Aplicada, Departamento de Medicina y Cirugía, Universidad Rovira i Virgili (URV), Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, España
| | - Teresa Auguet
- Grupo de recerca GEMMAIR (AGAUR)-Medicina Aplicada, Departamento de Medicina y Cirugía, Universidad Rovira i Virgili (URV), Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, España; Servicio de Medicina Interna, Hospital Universitario Joan XXIII, Tarragona, España.
| |
Collapse
|
27
|
Liao BM, Raddatz K, Zhong L, Parker BL, Raftery MJ, Schmitz-Peiffer C. Proteomic analysis of livers from fat-fed mice deficient in either PKCδ or PKCε identifies Htatip2 as a regulator of lipid metabolism. Proteomics 2014; 14:2578-87. [PMID: 25175814 DOI: 10.1002/pmic.201400202] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 08/13/2014] [Accepted: 08/27/2014] [Indexed: 11/08/2022]
Abstract
Insulin resistance contributes to the development of Type 2 diabetes, and is associated with lipid oversupply. Deletion of isoforms of the lipid-activated protein kinase C (PKC) family, PKCδ or PKCε, improves insulin action in fat-fed mice, but differentially affects hepatic lipid metabolism. To investigate the mechanisms involved, we employed an in vivo adaptation of SILAC to examine the effects of a fat diet together with deletion of PKCδ or PKCε on the expression of liver proteins. We identified a total of 3359 and 3488 proteins from the PKCδ and PKCε knockout study groups, respectively, and showed that several enzymes of lipid metabolism were affected by the fat diet. In fat-fed mice, 23 proteins showed changes upon PKCδ deletion while 19 proteins were affected by PKCε deletion. Enzymes of retinol metabolism were affected by the absence of either PKC. Pathway analysis indicated that monosaccharide metabolism was affected only upon PKCδ deletion, while isoprenoid biosynthesis was affected in a PKCε-specific manner. Certain proteins were regulated inversely, including HIV-1 tat interactive protein 2 (Htatip2). Overexpression or knockdown of Htatip2 in hepatocytes affected fatty acid storage and oxidation, consistent with a novel role in mediating the differential effects of PKC isoforms on lipid metabolism. All MS data have been deposited in the ProteomeXchange with identifier PXD000971 (http://proteomecentral.proteomexchange.org/dataset/PXD000971).
Collapse
Affiliation(s)
- Bing M Liao
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | | | | | | | | | | |
Collapse
|
28
|
Zhang J, Burrington CM, Davenport SK, Johnson AK, Horsman MJ, Chowdhry S, Greene MW. PKCδ regulates hepatic triglyceride accumulation and insulin signaling in Lepr(db/db) mice. Biochem Biophys Res Commun 2014; 450:1619-25. [PMID: 25035929 DOI: 10.1016/j.bbrc.2014.07.048] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 07/09/2014] [Indexed: 01/08/2023]
Abstract
PKCδ has been linked to key pathophysiological features of non-alcoholic fatty liver disease (NAFLD). Yet, our knowledge of PKCδ's role in NAFLD development and progression in obese models is limited. PKCδ(-/-)/Lepr(db)(/)(db) mice were generated to evaluate key pathophysiological features of NAFLD in mice. Hepatic histology, oxidative stress, apoptosis, gene expression, insulin signaling, and serum parameters were analyzed in Lepr(db)(/)(db) and PKCδ(-/-)/Lepr(db)(/)(db) mice. The absence of PKCδ did not abrogate the development of obesity in Lepr(db)(/)(db) mice. In contrast, serum triglyceride levels and epididymal white adipose tissue weight normalized to body weight were reduced in PKCδ(-/-)/Lepr(db)(/)(db) mice compared Lepr(db)(/)(db) mice. Analysis of insulin signaling in mice revealed that hepatic Akt and GSK3β phosphorylation were strongly stimulated by insulin in PKCδ(-/-)/Lepr(db)(/)(db) compared Lepr(db)(/)(db) mice. PKCδ may be involved in the development of obesity-associated NAFLD by regulating hepatic lipid metabolism and insulin signaling.
Collapse
Affiliation(s)
- Jian Zhang
- Boshell Diabetes and Metabolic Disease Research Program, Auburn University, Auburn, AL 36849, United States; College of Human Sciences, Auburn University, Auburn, AL 36849, United States
| | - Christine M Burrington
- Bassett Research Institute, Bassett Medical Center, Bassett Healthcare Network, Cooperstown, NY 13326, United States
| | - Samantha K Davenport
- Department of Pathology, Bassett Medical Center, Bassett Healthcare Network, Cooperstown, NY 13326, United States
| | - Andrew K Johnson
- Bassett Research Institute, Bassett Medical Center, Bassett Healthcare Network, Cooperstown, NY 13326, United States
| | - Melissa J Horsman
- Bassett Research Institute, Bassett Medical Center, Bassett Healthcare Network, Cooperstown, NY 13326, United States
| | - Saleem Chowdhry
- Department of Internal Medicine, Bassett Medical Center, Bassett Healthcare Network, Cooperstown, NY 13326, United States
| | - Michael W Greene
- Boshell Diabetes and Metabolic Disease Research Program, Auburn University, Auburn, AL 36849, United States; College of Human Sciences, Auburn University, Auburn, AL 36849, United States; Bassett Research Institute, Bassett Medical Center, Bassett Healthcare Network, Cooperstown, NY 13326, United States.
| |
Collapse
|
29
|
Hall AM, Soufi N, Chambers KT, Chen Z, Schweitzer GG, McCommis KS, Erion DM, Graham MJ, Su X, Finck BN. Abrogating monoacylglycerol acyltransferase activity in liver improves glucose tolerance and hepatic insulin signaling in obese mice. Diabetes 2014; 63:2284-96. [PMID: 24595352 PMCID: PMC4066334 DOI: 10.2337/db13-1502] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Monoacylglycerol acyltransferase (MGAT) enzymes convert monoacylglycerol to diacylglycerol (DAG), a lipid that has been linked to the development of hepatic insulin resistance through activation of protein kinase C (PKC). The expression of genes that encode MGAT enzymes is induced in the livers of insulin-resistant human subjects with nonalcoholic fatty liver disease, but whether MGAT activation is causal of hepatic steatosis or insulin resistance is unknown. We show that the expression of Mogat1, which encodes MGAT1, and MGAT activity are also increased in diet-induced obese (DIO) and ob/obmice. To probe the metabolic effects of MGAT1 in the livers of obese mice, we administered antisense oligonucleotides (ASOs) against Mogat1 to DIO and ob/ob mice for 3 weeks. Knockdown of Mogat1 in liver, which reduced hepatic MGAT activity, did not affect hepatic triacylglycerol content and unexpectedly increased total DAG content. Mogat1 inhibition also increased both membrane and cytosolic compartment DAG levels. However, Mogat1 ASO treatment significantly improved glucose tolerance and hepatic insulin signaling in obese mice. In summary, inactivation of hepatic MGAT activity, which is markedly increased in obese mice, improved glucose tolerance and hepatic insulin signaling independent of changes in body weight, intrahepatic DAG and TAG content, and PKC signaling.
Collapse
Affiliation(s)
- Angela M Hall
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
| | - Nisreen Soufi
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
| | - Kari T Chambers
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
| | - Zhouji Chen
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
| | - George G Schweitzer
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
| | - Kyle S McCommis
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
| | - Derek M Erion
- Cardiovascular, Metabolic, and Endocrine Diseases Research Unit, Pfizer Global Research and Development, Cambridge, MA
| | | | - Xiong Su
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MODepartment of Biochemistry and Molecular Biology, Medical College of Soochow University, Suzhou, China
| | - Brian N Finck
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
30
|
Tkachuk VA, Vorotnikov AV. Molecular Mechanisms of Insulin Resistance Development. DIABETES MELLITUS 2014. [DOI: 10.14341/dm2014229-40] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Insulin resistance (IR) is a phenomenon associated with an impaired ability of insulin to stimulate glucose uptake by target cells and to reduce the blood glucose level. A response increase in insulin secretion by the pancreas and hyperinsulinemia are compensatory reactions of the body. The development of IR leads to the inability of target cells to respond to insulin that results in developing type 2 diabetes mellitus (T2DM) and metabolic syndrome. For this reason, the metabolic syndrome is defined in practice as a combination of IR with one or more pathologies such as T2DM, arterial hypertension, dyslipidemia, abdominal obesity, non-alcoholic fatty liver disease, and some others. However, a combination of high blood glucose and insulin levels always serves as its physiological criterion. IR should be considered as a systemic failure of the endocrine regulation in the body. Physiological causes of IR are diverse. The main ones are nutritional overload and accumulation of certain lipids and their metabolites in cells, low physical activity, chronic inflammation and stress of various nature, including oxidative and endoplasmic reticulum stress (impairment of damaged protein degradation in the cell). Recent studies have demonstrated that these physiological mechanisms likely act through a single intracellular scenario. This is the impairment of signal transduction from the insulin receptor to its targets via the negative feedback mechanism in intracellular insulin-dependent signaling cascades. This review describes the physiological and intracellular mechanisms of insulin action and focuses on their abnormalities upon IR development. Finally, feasible trends in early molecular diagnosis and therapy of IR are discussed.
Collapse
|
31
|
Birkenfeld AL, Shulman GI. Nonalcoholic fatty liver disease, hepatic insulin resistance, and type 2 diabetes. Hepatology 2014; 59:713-23. [PMID: 23929732 PMCID: PMC3946772 DOI: 10.1002/hep.26672] [Citation(s) in RCA: 522] [Impact Index Per Article: 52.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 07/31/2013] [Indexed: 12/12/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD), hepatic insulin resistance, and type 2 diabetes are all strongly associated and are all reaching epidemic proportions. Whether there is a causal link between NAFLD and hepatic insulin resistance is controversial. This review will discuss recent studies in both humans and animal models of NAFLD that have implicated increases in hepatic diacylglycerol (DAG) content leading to activation of novel protein kinase Cϵ (PKCϵ) resulting in decreased insulin signaling in the pathogenesis of NAFLD-associated hepatic insulin resistance and type 2 diabetes. The DAG-PKCϵ hypothesis can explain the occurrence of hepatic insulin resistance observed in most cases of NAFLD associated with obesity, lipodystrophy, and type 2 diabetes.
Collapse
Affiliation(s)
- Andreas L. Birkenfeld
- Charité - University School of Medicine, Department of Endocrinology Diabetes and Nutrition, Center for Cardiovascular Research, Berlin, Germany
- Howard Hughes Medical Institute and the Departments of Internal Medicine and Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Gerald I. Shulman
- Howard Hughes Medical Institute and the Departments of Internal Medicine and Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
32
|
Greene MW, Burrington CM, Lynch DT, Davenport SK, Johnson AK, Horsman MJ, Chowdhry S, Zhang J, Sparks JD, Tirrell PC. Lipid metabolism, oxidative stress and cell death are regulated by PKC delta in a dietary model of nonalcoholic steatohepatitis. PLoS One 2014; 9:e85848. [PMID: 24454937 PMCID: PMC3893275 DOI: 10.1371/journal.pone.0085848] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 12/03/2013] [Indexed: 12/31/2022] Open
Abstract
Steatosis, oxidative stress, and apoptosis underlie the development of nonalcoholic steatohepatitis (NASH). Protein kinase C delta (PKCδ) has been implicated in fatty liver disease and is activated in the methionine and choline-deficient (MCD) diet model of NASH, yet its pathophysiological importance towards steatohepatitis progression is uncertain. We therefore addressed the role of PKCδ in the development of steatosis, inflammation, oxidative stress, apoptosis, and fibrosis in an animal model of NASH. We fed PKCδ−/− mice and wildtype littermates a control or MCD diet. PKCδ−/− primary hepatocytes were used to evaluate the direct effects of fatty acids on hepatocyte lipid metabolism gene expression. A reduction in hepatic steatosis and triglyceride levels were observed between wildtype and PKCδ−/− mice fed the MCD diet. The hepatic expression of key regulators of β-oxidation and plasma triglyceride metabolism was significantly reduced in PKCδ−/− mice and changes in serum triglyceride were blocked in PKCδ−/− mice. MCD diet-induced hepatic oxidative stress and hepatocyte apoptosis were reduced in PKCδ−/− mice. MCD diet-induced NADPH oxidase activity and p47phox membrane translocation were blunted and blocked, respectively, in PKCδ−/− mice. Expression of pro-apoptotic genes and caspase 3 and 9 cleavage in the liver of MCD diet fed PKCδ−/− mice were blunted and blocked, respectively. Surprisingly, no differences in MCD diet-induced fibrosis or pro-fibrotic gene expression were observed in 8 week MCD diet fed PKCδ−/− mice. Our results suggest that PKCδ plays a role in key pathological features of fatty liver disease but not ultimately in fibrosis in the MCD diet model of NASH.
Collapse
Affiliation(s)
- Michael W. Greene
- Boshell Diabetes and Metabolic Disease Research Program, Auburn University, Auburn, Alabama, United States of America
- College of Human Sciences, Auburn University, Auburn, Alabama, United States of America
- Bassett Research Institute, Bassett Medical Center, Bassett Healthcare Network, Cooperstown, New York, United States of America
- * E-mail:
| | - Christine M. Burrington
- Bassett Research Institute, Bassett Medical Center, Bassett Healthcare Network, Cooperstown, New York, United States of America
| | - Darin T. Lynch
- Bassett Research Institute, Bassett Medical Center, Bassett Healthcare Network, Cooperstown, New York, United States of America
| | - Samantha K. Davenport
- Department of Pathology, Bassett Medical Center, Bassett Healthcare Network, Cooperstown, New York, United States of America
| | - Andrew K. Johnson
- Bassett Research Institute, Bassett Medical Center, Bassett Healthcare Network, Cooperstown, New York, United States of America
| | - Melissa J. Horsman
- Bassett Research Institute, Bassett Medical Center, Bassett Healthcare Network, Cooperstown, New York, United States of America
| | - Saleem Chowdhry
- Department of Internal Medicine, Bassett Medical Center, Bassett Healthcare Network, Cooperstown, New York, United States of America
| | - Jian Zhang
- College of Human Sciences, Auburn University, Auburn, Alabama, United States of America
| | - Janet D. Sparks
- University of Rochester Medical Center, School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Paul C. Tirrell
- Department of Internal Medicine, Bassett Medical Center, Bassett Healthcare Network, Cooperstown, New York, United States of America
| |
Collapse
|
33
|
Greene MW, Burrington CM, Luo Y, Ruhoff MS, Lynch DT, Chaithongdi N. PKCδ is activated in the liver of obese Zucker rats and mediates diet-induced whole body insulin resistance and hepatocyte cellular insulin resistance. J Nutr Biochem 2013; 25:281-8. [PMID: 24524901 DOI: 10.1016/j.jnutbio.2013.10.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 09/06/2013] [Accepted: 10/08/2013] [Indexed: 02/06/2023]
Abstract
Insulin resistance can arise when pathological levels of free fatty acids (FFAs) and proinflammatory cytokines disrupt insulin signaling. Protein kinase C delta (PKCδ) is a FFA- and a proinflammatory cytokine-regulated protein kinase that is associated with inhibition of insulin signaling and action. To gain insight into the role of PKCδ in insulin resistance, PKCδ activation was studied in a genetic model of obesity-linked insulin resistance. PKCδ was found to be activated in the liver of obese insulin-resistant Zucker rats and in isolated cultured hepatocytes. PKCδ was further studied in PKCδ-null mice and their wild-type littermates fed a high-fat or control diet for 10 weeks. PKCδ-null mice on a high-fat diet had improved insulin sensitivity and hepatic insulin signaling compared to wild-type littermates. Additionally, the deleterious effect of a high-fat diet on glucose tolerance in wild-type mice was completely blocked in PKCδ-null mice. To directly test the role of PKCδ in cellular insulin resistance, primary hepatocytes from the high-fat diet mice were isolated and stimulated with insulin. Primary hepatocytes from PKCδ-null mice had improved insulin-stimulated Akt and FOXO phosphorylation compared to hepatocytes from wild-type littermates. Consistent with this result, tumor necrosis factor alpha-mediated inhibition of insulin signaling was blocked in PKCδ knockdown primary hepatocytes. These results indicate that PKCδ plays a role in insulin resistance and is consistent with the hypothesis that PKCδ is a negative regulator of insulin signaling and thus may be a therapeutic target for the treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Michael W Greene
- Boshell Metabolic Diseases and Diabetes Program, Auburn University, Auburn, AL 36849, USA; College of Human Sciences, Auburn University, Auburn, AL 36849, USA; Bassett Research Institute, Bassett Healthcare Network, Cooperstown, NY, USA.
| | | | - Yuwen Luo
- College of Human Sciences, Auburn University, Auburn, AL 36849, USA
| | - Mary S Ruhoff
- Bassett Research Institute, Bassett Healthcare Network, Cooperstown, NY, USA
| | - Darin T Lynch
- Bassett Research Institute, Bassett Healthcare Network, Cooperstown, NY, USA
| | - Niyutchai Chaithongdi
- Department of Internal Medicine, Bassett Medical Center, Bassett Healthcare Network, Cooperstown, NY, USA
| |
Collapse
|
34
|
Schmitz-Peiffer C. The tail wagging the dog--regulation of lipid metabolism by protein kinase C. FEBS J 2013; 280:5371-83. [PMID: 23587021 DOI: 10.1111/febs.12285] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2013] [Revised: 03/19/2013] [Accepted: 04/11/2013] [Indexed: 12/12/2022]
Abstract
Upon their discovery almost 40 years ago, isoforms of the lipid-activated protein kinase C (PKC) family were initially regarded only as downstream effectors of the second messengers calcium and diacylglycerol, undergoing activation upon phospholipid hydrolysis in response to acute stimuli. Subsequently, several isoforms were found to be associated with the inhibitory effects of lipid over-supply on glucose homeostasis, especially the negative cross-talk with insulin signal transduction, observed upon accumulation of diacylglycerol in insulin target tissues. The PKC family has therefore attracted much attention in diabetes and obesity research, because intracellular lipid accumulation is strongly correlated with defective insulin action and the development of type 2 diabetes. Causal roles for various isoforms in the generation of insulin resistance have more recently been confirmed using PKC-deficient mice. However, during characterization of these animals, it became increasingly evident that the enzymes play key roles in the modulation of lipid metabolism itself, and may control the supply of lipids between tissues such as adipose and liver. Molecular studies have also demonstrated roles for PKC isoforms in several aspects of lipid metabolism, such as adipocyte differentiation and hepatic lipogenesis. While the precise mechanisms involved, especially the identities of protein substrates, are still unclear, the emerging picture suggests that the currently held view of the contribution of PKC isoforms to metabolism is an over-simplification. Although PKCs may inhibit insulin signal transduction, these enzymes are not merely downstream effectors of lipid accumulation, but in fact control the fate of fatty acids, thus the tail wags the dog.
Collapse
Affiliation(s)
- Carsten Schmitz-Peiffer
- Diabetes and Obesity Program, Garvan Institute of Medical Research, Sydney, NSW, Australia; St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
35
|
Gong J, Hoyos B, Acin-Perez R, Vinogradov V, Shabrova E, Zhao F, Leitges M, Fischman D, Manfredi G, Hammerling U. Two protein kinase C isoforms, δ and ε, regulate energy homeostasis in mitochondria by transmitting opposing signals to the pyruvate dehydrogenase complex. FASEB J 2012; 26:3537-49. [PMID: 22573912 DOI: 10.1096/fj.11-197376] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Energy production in mitochondria is a multistep process that requires coordination of several subsystems. While reversible phosphorylation is emerging as the principal tool, it is still unclear how this signal network senses the workloads of processes as different as fuel procurement, catabolism in the Krebs cycle, and stepwise oxidation of reducing equivalents in the electron transfer chain. We previously proposed that mitochondria use oxidized cytochrome c in concert with retinol to activate protein kinase Cδ, thereby linking a prominent kinase network to the redox balance of the ETC. Here, we show that activation of PKCε in mitochondria also requires retinol as a cofactor, implying a redox-mechanism. Whereas activated PKCδ transmits a stimulatory signal to the pyruvate dehdyrogenase complex (PDHC), PKCε opposes this signal and inhibits the PDHC. Our results suggest that the balance between PKCδ and ε is of paramount importance not only for flux of fuel entering the Krebs cycle but for overall energy homeostasis. We observed that the synthetic retinoid fenretinide substituted for the retinol cofactor function but, on chronic use, distorted this signal balance, leading to predominance of PKCε over PKCδ. The suppression of the PDHC might explain the proapoptotic effect of fenretinide on tumor cells, as well as the diminished adiposity observed in experimental animals and humans. Furthermore, a disturbed balance between PKCδ and PKCε might underlie the injury inflicted on the ischemic myocardium during reperfusion. dehydrogenase complex.
Collapse
Affiliation(s)
- Jianli Gong
- Immunology Program, Sloan-Kettering Institute for Cancer Research, New York, New York 10065, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Nonalcoholic fatty liver disease (NAFLD) is now the most frequent chronic liver disease in Western societies, affecting one in four adults in the USA, and is strongly associated with hepatic insulin resistance, a major risk factor in the pathogenesis of type 2 diabetes. Although the cellular mechanisms underlying this relationship are unknown, hepatic accumulation of diacylglycerol (DAG) in both animals and humans has been linked to hepatic insulin resistance. In this Perspective, we discuss the role of DAG activation of protein kinase Cε as the mechanism responsible for NAFLD-associated hepatic insulin resistance seen in obesity, type 2 diabetes, and lipodystrophy.
Collapse
Affiliation(s)
- François R Jornayvaz
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
| | | |
Collapse
|
37
|
Prentki M, Madiraju SRM. Glycerolipid/free fatty acid cycle and islet β-cell function in health, obesity and diabetes. Mol Cell Endocrinol 2012; 353:88-100. [PMID: 22108437 DOI: 10.1016/j.mce.2011.11.004] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Revised: 11/07/2011] [Accepted: 11/07/2011] [Indexed: 12/16/2022]
Abstract
Pancreatic β-cells secrete insulin in response to fluctuations in blood fuel concentrations, in particular glucose and fatty acids. However, chronic fuel surfeit can overwhelm the metabolic, signaling and secretory capacity of the β-cell leading to its dysfunction and death - often referred to as glucolipotoxicity. In β-cells and many other cells, glucose and lipid metabolic pathways converge into a glycerolipid/free fatty acid (GL/FFA) cycle, which is driven by the substrates, glycerol-3-phosphate and fatty acyl-CoA, derived from glucose and fatty acids, respectively. Although the overall operation of GL/FFA cycle, consisting of lipolysis and lipogenesis, is "futile" in terms of energy expenditure, this metabolic cycle likely plays an indispensable role for various β-cell functions, in particular insulin secretion and excess fuel detoxification. In this review, we discuss the significance of GL/FFA cycle in the β-cell, its regulation and role in generating essential metabolic signals that participate in the lipid amplification arm of glucose stimulated insulin secretion and in β-cell growth. We propose the novel concept that the lipolytic segment of GL/FFA cycle is instrumental in producing signals for insulin secretion, whereas, the lipogenic segment generates signals relevant for β-cell survival/death and growth/proliferation.
Collapse
Affiliation(s)
- Marc Prentki
- Departments of Nutrition and Biochemistry, University of Montreal, Montreal Diabetes Research Center, CR-CHUM, Technopôle Angus, 2901, Montreal, Canada QC H1W 4A4.
| | | |
Collapse
|
38
|
Samuel VT, Shulman GI. Mechanisms for insulin resistance: common threads and missing links. Cell 2012; 148:852-71. [PMID: 22385956 DOI: 10.1016/j.cell.2012.02.017] [Citation(s) in RCA: 1506] [Impact Index Per Article: 125.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Indexed: 02/07/2023]
Abstract
Insulin resistance is a complex metabolic disorder that defies explanation by a single etiological pathway. Accumulation of ectopic lipid metabolites, activation of the unfolded protein response (UPR) pathway, and innate immune pathways have all been implicated in the pathogenesis of insulin resistance. However, these pathways are also closely linked to changes in fatty acid uptake, lipogenesis, and energy expenditure that can impact ectopic lipid deposition. Ultimately, these cellular changes may converge to promote the accumulation of specific lipid metabolites (diacylglycerols and/or ceramides) in liver and skeletal muscle, a common final pathway leading to impaired insulin signaling and insulin resistance.
Collapse
Affiliation(s)
- Varman T Samuel
- Department of Medicine, Yale University School of Medicine, New Haven, CT 06510, USA.
| | | |
Collapse
|
39
|
Bradley SJ, Challiss RAJ. Defining protein kinase/phosphatase isoenzymic regulation of mGlu₅ receptor-stimulated phospholipase C and Ca²⁺ responses in astrocytes. Br J Pharmacol 2012; 164:755-71. [PMID: 21486279 DOI: 10.1111/j.1476-5381.2011.01421.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND AND PURPOSE Cyclical phosphorylation and dephosphorylation of a key residue within the C-terminal domain of the activated type 5 metabotropic glutamate (mGlu₅) receptor is believed to cause the synchronous, oscillatory changes in inositol 1,4,5-trisphosphate and Ca²⁺ levels observed in a variety of cell types. Here, we have attempted to better define the kinase and phosphatase enzymes involved in this modulation. EXPERIMENTAL APPROACH Ca²⁺ and [³H]inositol phosphate ([³H]IP(x) ) measurements in astrocyte preparations have been used to evaluate the effects of pharmacological inhibition of protein kinase C (PKC) and protein phosphatase activities and small interfering RNA-mediated specific PKC isoenzymic knock-down on mGlu₅ receptor signalling. KEY RESULTS Ca²⁺ oscillation frequency or [³H]IP(x) accumulation in astrocytes stimulated by mGlu₅ receptors, was concentration-dependently decreased by protein phosphatase-1/2A inhibition or by PKC activation. PKC inhibition also increased [³H]IP(x) accumulation two- to threefold and changed the Ca²⁺ response into a peak-plateau response. However, selective inhibition of conventional PKC isoenzymes or preventing changes in [Ca²⁺](i) concentration by BAPTA-AM loading was without effect on mGlu₅ receptor-stimulated [³H]IP(x) accumulation. Selective knock-down of PKCδ was without effect on glutamate-stimulated Ca²⁺ responses; however, selective PKCε knock-down in astrocytes changed Ca²⁺ responses from oscillatory into peak-plateau type. CONCLUSION AND IMPLICATIONS These data confirm the acute regulation of mGlu₅ receptor signalling by protein kinases and protein phosphatases and provide novel data pinpointing the isoenzymic dependence of this regulation in the native mGlu₅ receptor-expressing rat cortical astrocyte. These data also highlight a potential alternative mechanism by which mGlu₅ receptor signalling might be therapeutically manipulated.
Collapse
Affiliation(s)
- S J Bradley
- Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, UK
| | | |
Collapse
|
40
|
Shankar E, Vykhovanets EV, Vykhovanets OV, MacLennan GT, Singh R, Bhaskaran N, Shukla S, Gupta S. High-fat diet activates pro-inflammatory response in the prostate through association of Stat-3 and NF-κB. Prostate 2012; 72:233-43. [PMID: 21604287 PMCID: PMC3161175 DOI: 10.1002/pros.21425] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Accepted: 04/28/2011] [Indexed: 01/05/2023]
Abstract
BACKGROUND Signal transducer and activator of transcription (Stat)-3 and nuclear factor-kappa B (NF-κB) are important signaling pathways constitutively activated during inflammation. We previously reported that high-fat diet (HFD) intake induces oxidative stress in the prostate through elevated expression of NADPH oxidase subunits causing NF-κB activation. We sought to determine whether Stat-3 is involved in the activation of NF-κB in the prostate as a result of HFD feeding, leading to inflammation. METHODS C57BL/6 mice were either fed with regular diet (RD) or HFD for 4 and 8 weeks. Plasma cytokine levels were determined by multiplex analysis. Western blotting was performed to determine the expression of NF-κB, Stat-3, Akt, PDK1, PKCε, and their phosphorylated forms along with pathologic evaluation of the prostate. Immunoprecipitation and electrophoretic mobility shift assay (EMSA) were conducted to study the association between Stat-3 and NF-κB. RESULTS C57BL/6 mice fed with HFD showed a significant increase in the plasma levels of IL-1ß, IL-6, IL-17, and TNFα after 4 and 8 weeks of feeding, compared with RD controls. HFD feeding elevated the intraprostatic expression of IL-6 and caused activation of PKCε and Akt, the upstream kinase regulating Stat-3 and NF-κB. Nuclear extracts from the prostates of mice fed with HFD exhibited constitutively activated levels of Stat-3 and NF-κB/p65. Increased association between the activated forms of Stat-3 and NF-κB/p65 was observed in the nucleus as a result of HFD feeding, a finding that was accompanied by morphologic evidence of increased intraprostatic inflammation. CONCLUSIONS Our findings suggest that HFD activates Stat-3 and NF-κB/p65 in the prostate, and their interaction is associated with increased inflammation in the prostate.
Collapse
Affiliation(s)
- Eswar Shankar
- Department of Urology, Case Western Reserve University & The Urology Institute, University Hospitals Case Medical Center, Cleveland, Ohio 44106
| | - Eugene V Vykhovanets
- Department of Urology, Case Western Reserve University & The Urology Institute, University Hospitals Case Medical Center, Cleveland, Ohio 44106
| | - Olena V Vykhovanets
- Department of Urology, Case Western Reserve University & The Urology Institute, University Hospitals Case Medical Center, Cleveland, Ohio 44106
| | - Gregory T MacLennan
- Department of Pathology, Case Western Reserve University & The Urology Institute, University Hospitals Case Medical Center, Cleveland, Ohio 44106
| | - Rajesh Singh
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, Georgia 30310
| | - Natarajan Bhaskaran
- Department of Urology, Case Western Reserve University & The Urology Institute, University Hospitals Case Medical Center, Cleveland, Ohio 44106
| | - Sanjeev Shukla
- Department of Urology, Case Western Reserve University & The Urology Institute, University Hospitals Case Medical Center, Cleveland, Ohio 44106
| | - Sanjay Gupta
- Department of Urology, Case Western Reserve University & The Urology Institute, University Hospitals Case Medical Center, Cleveland, Ohio 44106
- Department of Nutrition, Case Western Reserve University & The Urology Institute, University Hospitals Case Medical Center, Cleveland, Ohio 44106
| |
Collapse
|
41
|
Kewalramani G, Fink LN, Asadi F, Klip A. Palmitate-activated macrophages confer insulin resistance to muscle cells by a mechanism involving protein kinase C θ and ε. PLoS One 2011; 6:e26947. [PMID: 22046423 PMCID: PMC3202600 DOI: 10.1371/journal.pone.0026947] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Accepted: 10/06/2011] [Indexed: 12/16/2022] Open
Abstract
Background Macrophage-derived factors contribute to whole-body insulin resistance, partly by impinging on metabolically active tissues. As proof of principle for this interaction, conditioned medium from macrophages treated with palmitate (CM-PA) reduces insulin action and glucose uptake in muscle cells. However, the mechanism whereby CM-PA confers this negative response onto muscle cells remains unknown. Methodology/Principal Findings L6-GLUT4myc myoblasts were exposed for 24 h to palmitate-free conditioned medium from RAW 264.7 macrophages pre-treated with 0.5 mM palmitate for 6 h. This palmitate-free CM-PA, containing selective cytokines and chemokines, inhibited myoblast insulin-stimulated insulin receptor substrate 1 (IRS1) tyrosine phosphorylation, AS160 phosphorylation, GLUT4 translocation and glucose uptake. These effects were accompanied by a rise in c-Jun N-terminal kinase (JNK) activation, degradation of Inhibitor of κBα (IκBα), and elevated expression of proinflammatory cytokines in myoblasts. Notably, CM-PA caused IRS1 phosphorylation on Ser1101, and phosphorylation of novel PKCθ and ε. Co-incubation of myoblasts with CM-PA and the novel and conventional PKC inhibitor Gö6983 (but not with the conventional PKC inhibitor Gö6976) prevented PKCθ and ε activation, JNK phosphorylation, restored IκBα mass and reduced proinflammatory cytokine production. Gö6983 also restored insulin signalling and glucose uptake in myoblasts. Moreover, co-silencing both novel PKC θ and ε isoforms in myoblasts by RNA interference, but not their individual silencing, prevented the inflammatory response and restored insulin sensitivity to CM-PA-treated myoblasts. Conclusions/Clinical Significance The results suggest that the block in muscle insulin action caused by CM-PA is mediated by novel PKCθ and PKCε. This study re-establishes the participation of macrophages as a relay in the action of fatty acids on muscle cells, and further identifies PKCθ and PKCε as key elements in the inflammatory and insulin resistance responses of muscle cells to macrophage products. Furthermore, it portrays these PKC isoforms as potential targets for the treatment of fatty acid-induced, inflammation-linked insulin resistance.
Collapse
Affiliation(s)
| | - Lisbeth Nielsen Fink
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
- Hagedorn Research Institute, Novo Nordisk A/S, Gentofte, Denmark
| | - Farzad Asadi
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
- Department of Biochemistry, School of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Amira Klip
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
- * E-mail:
| |
Collapse
|
42
|
Raddatz K, Turner N, Frangioudakis G, Liao BM, Pedersen DJ, Cantley J, Wilks D, Preston E, Hegarty BD, Leitges M, Raftery MJ, Biden TJ, Schmitz-Peiffer C. Time-dependent effects of Prkce deletion on glucose homeostasis and hepatic lipid metabolism on dietary lipid oversupply in mice. Diabetologia 2011; 54:1447-56. [PMID: 21347625 DOI: 10.1007/s00125-011-2073-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2010] [Accepted: 01/10/2011] [Indexed: 02/06/2023]
Abstract
AIMS/HYPOTHESIS We examined the time-dependent effects of deletion of the gene encoding protein kinase C epsilon (Prkce) on glucose homeostasis, insulin secretion and hepatic lipid metabolism in fat-fed mice. METHODS Prkce(-/-) and wild-type (WT) mice were fed a high-fat diet for 1 to 16 weeks and subjected to i.p. glucose tolerance tests (ipGTT) and indirect calorimetry. We also investigated gene expression and protein levels by RT-PCR, quantitative protein profiling (isobaric tag for relative and absolute quantification; iTRAQ) and immunoblotting. Lipid levels, mitochondrial oxidative capacity and lipid metabolism were assessed in liver and primary hepatocytes. RESULTS While fat-fed WT mice became glucose intolerant after 1 week, Prkce(-/-) mice exhibited normal glucose and insulin levels. iTRAQ suggested differences in lipid metabolism and oxidative phosphorylation between fat-fed WT and Prkce(-/-) animals. Liver triacylglycerols were increased in fat-fed Prkce(-/-) mice, resulting from altered lipid partitioning which promoted esterification of fatty acids in hepatocytes. In WT mice, fat feeding elevated oxygen consumption in vivo and in isolated liver mitochondria, but these increases were not seen in Prkce(-/-) mice. Prkce(-/-) hepatocytes also exhibited reduced production of reactive oxygen species (ROS) in the presence of palmitate. After 16 weeks of fat feeding, however, the improved glucose tolerance in fat-fed Prkce(-/-) mice was instead associated with increased insulin secretion during ipGTT, as we have previously reported. CONCLUSIONS/INTERPRETATION Prkce deletion ameliorates diet-induced glucose intolerance via two temporally distinct phenotypes. Protection against insulin resistance is associated with changes in hepatic lipid partitioning, which may reduce the acute inhibitory effects of fatty acid catabolism, such as ROS generation. In the longer term, enhancement of glucose-stimulated insulin secretion prevails.
Collapse
Affiliation(s)
- K Raddatz
- Garvan Institute of Medical Research, 384 Victoria Street, Sydney, NSW 2010, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Bezy O, Tran TT, Pihlajamäki J, Suzuki R, Emanuelli B, Winnay J, Mori MA, Haas J, Biddinger SB, Leitges M, Goldfine AB, Patti ME, King GL, Kahn CR. PKCδ regulates hepatic insulin sensitivity and hepatosteatosis in mice and humans. J Clin Invest 2011; 121:2504-17. [PMID: 21576825 DOI: 10.1172/jci46045] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2010] [Accepted: 03/30/2011] [Indexed: 12/27/2022] Open
Abstract
C57BL/6J and 129S6/Sv (B6 and 129) mice differ dramatically in their susceptibility to developing diabetes in response to diet- or genetically induced insulin resistance. A major locus contributing to this difference has been mapped to a region on mouse chromosome 14 that contains the gene encoding PKCδ. Here, we found that PKCδ expression in liver was 2-fold higher in B6 versus 129 mice from birth and was further increased in B6 but not 129 mice in response to a high-fat diet. PRKCD gene expression was also elevated in obese humans and was positively correlated with fasting glucose and circulating triglycerides. Mice with global or liver-specific inactivation of the Prkcd gene displayed increased hepatic insulin signaling and reduced expression of gluconeogenic and lipogenic enzymes. This resulted in increased insulin-induced suppression of hepatic gluconeogenesis, improved glucose tolerance, and reduced hepatosteatosis with aging. Conversely, mice with liver-specific overexpression of PKCδ developed hepatic insulin resistance characterized by decreased insulin signaling, enhanced lipogenic gene expression, and hepatosteatosis. Therefore, changes in the expression and regulation of PKCδ between strains of mice and in obese humans play an important role in the genetic risk of hepatic insulin resistance, glucose intolerance, and hepatosteatosis; and thus PKCδ may be a potential target in the treatment of metabolic syndrome.
Collapse
Affiliation(s)
- Olivier Bezy
- Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02215, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Cantley J, Boslem E, Laybutt DR, Cordery DV, Pearson G, Carpenter L, Leitges M, Biden TJ. Deletion of protein kinase Cδ in mice modulates stability of inflammatory genes and protects against cytokine-stimulated beta cell death in vitro and in vivo. Diabetologia 2011; 54:380-9. [PMID: 21103982 DOI: 10.1007/s00125-010-1962-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2010] [Accepted: 10/06/2010] [Indexed: 12/23/2022]
Abstract
AIMS/HYPOTHESIS Proinflammatory cytokines contribute to beta cell destruction in type 1 diabetes, but the mechanisms are incompletely understood. The aim of the current study was to address the role of the protein kinase C (PKC) isoform PKCδ, a diverse regulator of cell death, in cytokine-stimulated apoptosis in primary beta cells. METHODS Islets isolated from wild-type or Prkcd(-/-) mice were treated with IL-1β, TNF-α and IFNγ and assayed for apoptosis, nitric oxide (NO) generation and insulin secretion. Activation of signalling pathways, apoptosis and endoplasmic reticulum (ER) stress were determined by immunoblotting. Stabilisation of mRNA transcripts was measured by RT-PCR following transcriptional arrest. Mice were injected with multiple low doses of streptozotocin (MLD-STZ) and fasting blood glucose monitored. RESULTS Deletion of Prkcd inhibited apoptosis and NO generation in islets stimulated ex vivo with cytokines. It also delayed the onset of hyperglycaemia in MLD-STZ-treated mice. Activation of ERK, p38, JNK, AKT1, the ER stress markers DDIT3 and phospho-EIF2α and the intrinsic apoptotic markers BCL2 and MCL1 was not different between genotypes. However, deletion of Prkcd destabilised mRNA transcripts for Nos2, and for multiple components of the toll-like receptor 2 (TLR2) signalling complex, which resulted in disrupted TLR2 signalling. CONCLUSIONS/INTERPRETATION Loss of PKCδ partially protects against hyperglycaemia in the MLD-STZ model in vivo, and against cytokine-mediated apoptosis in vitro. This is accompanied by reduced NO generation and destabilisation of Nos2 and components of the TLR2 signalling pathway. The results highlight a mechanism for regulating proinflammatory gene expression in beta cells independently of transcription.
Collapse
Affiliation(s)
- J Cantley
- Garvan Institute of Medical Research, St Vincent's Hospital, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Greene MW, Burrington CM, Ruhoff MS, Johnson AK, Chongkrairatanakul T, Kangwanpornsiri A. PKC{delta} is activated in a dietary model of steatohepatitis and regulates endoplasmic reticulum stress and cell death. J Biol Chem 2010; 285:42115-29. [PMID: 20971848 DOI: 10.1074/jbc.m110.168575] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Hepatic steatosis can progress to the clinical condition of non-alcoholic steatohepatitis (NASH), which is a precursor of more serious liver diseases. The novel PKC isoforms δ and ε are activated by lipid metabolites and have been implicated in lipid-induced hepatic disease. Using a methionine- and choline-deficient (MCD) dietary model of NASH, we addressed the question of whether hepatic PKCδ and PKCε are activated. With progression from steatosis to steatohepatitis, there was activation and increased PKCδ protein content coincident with hepatic endoplasmic reticulum (ER) stress parameters. To examine whether similar changes could be induced in vitro, McA-RH 7777 (McA) hepatoma cells were used. We observed that McA cells stored triglyceride and released alanine aminotransferase (ALT) when treated with MCD medium in the presence of fatty acids. Further, MCD medium with palmitic acid, but not oleic or linoleic acids, maximally activated PKCδ and stimulated ER stress. In PKCδ knockdown McA cells, MCD/fatty acid medium-induced ALT release and ER stress induction were completely blocked, but triglyceride storage was not. In addition, a reduction in the uptake of propidium iodide and the number of apoptotic nuclei and a significant increase in cell viability and DNA content were observed in PKCδ knockdown McA cells incubated in MCD medium with palmitic acid. Our studies show that PKCδ activation and protein levels are elevated in an animal model of steatohepatitis, which was recapitulated in a cell model, supporting the conclusion that PKCδ plays a role in ALT release, the ER stress signal, and cell death.
Collapse
Affiliation(s)
- Michael W Greene
- Bassett Research Institute, Bassett Medical Center, Bassett Healthcare Network, Cooperstown, New York 13326, USA.
| | | | | | | | | | | |
Collapse
|
46
|
Wallerstedt E, Smith U, Andersson CX. Protein kinase C-delta is involved in the inflammatory effect of IL-6 in mouse adipose cells. Diabetologia 2010; 53:946-54. [PMID: 20151299 DOI: 10.1007/s00125-010-1668-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2009] [Accepted: 12/17/2009] [Indexed: 01/01/2023]
Abstract
AIMS/HYPOTHESIS The aim of the study was to address the role of protein kinase C-delta (PKCdelta) on phosphorylation of signal transducer and activator of transcription 3 (STAT3) and activation of inflammatory genes in response to IL-6 in adipose cells. METHODS Differentiated mouse 3T3-L1 adipocytes preincubated with the PKCdelta inhibitor rottlerin and mouse embryonic fibroblasts (MEFs) lacking PKCdelta were incubated with IL-6 and/or insulin. RNA was extracted and the gene expression was analysed by real-time PCR, while the proteins from total, nuclear and cytoplasmic lysates were analysed by immunoblotting. RESULTS Inhibition of PKCdelta by rottlerin significantly reduced both Ser-727 and Tyr-705 phosphorylation of STAT3. Consequently, nuclear translocation of STAT3 and the IL-6-induced gene transcription and protein release of the inflammatory molecule serum amyloid A 3 (SAA3) were reduced. Similarly, the IL-6-regulated gene transcription of Il-6 (also known as Il6) to Hp and the feedback inhibitor of IL-6, Socs3, were also attenuated by rottlerin. Furthermore, PKCdelta was found to translocate to the nucleus following IL-6 treatment and this was also reduced by rottlerin. In agreement with the effect of rottlerin, Pkcdelta (also known as Prkcd) ( -/- ) MEFs also displayed a markedly reduced ability of IL-6 to activate the transcription of Saa3, Hp, Socs3 and Il6 genes compared with wild-type MEFs. These results correlated with a reduced nuclear translocation and phosphorylation of STAT3. CONCLUSIONS/INTERPRETATION These results show that PKCdelta plays a key role in the inflammatory effect of IL-6 in adipose cells and may be a suitable target for novel anti-inflammatory agents.
Collapse
Affiliation(s)
- E Wallerstedt
- The Lundberg Laboratory for Diabetes Research, Center of Excellence for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine/Diabetes, The Sahlgrenska Academy at University of Gothenburg, Blå Stråket 5, SE-413 45, Gothenburg, Sweden
| | | | | |
Collapse
|
47
|
Current literature in diabetes. Diabetes Metab Res Rev 2010; 26:i-xi. [PMID: 20474064 DOI: 10.1002/dmrr.1019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
48
|
Bikman BT, Zheng D, Kane DA, Anderson EJ, Woodlief TL, Price JW, Dohm GL, Neufer PD, Cortright RN. Metformin Improves Insulin Signaling in Obese Rats via Reduced IKKbeta Action in a Fiber-Type Specific Manner. J Obes 2010; 2010:970865. [PMID: 20798864 PMCID: PMC2925476 DOI: 10.1155/2010/970865] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2009] [Accepted: 10/27/2009] [Indexed: 02/06/2023] Open
Abstract
Metformin is a widely used insulin-sensitizing drug, though its mechanisms are not fully understood. Metformin has been shown to activate AMPK in skeletal muscle; however, its effects on the inhibitor of kappaB kinasebeta (IKKbeta) in this same tissue are unknown. The aim of this study was to (1) determine the ability of metformin to attenuate IKKbeta action, (2) determine whether changes in AMPK activity are associated with changes in IKKbeta action in skeletal muscle, and (3) examine whether changes in AMPK and IKKbeta function are consistent with improved insulin signaling. Lean and obese male Zuckers received either vehicle or metformin by oral gavage daily for four weeks (four groups of eight). Proteins were measured in white gastrocnemius (WG), red gastrocnemius (RG), and soleus. AMPK phosphorylation increased (P < .05) in WG in both lean (57%) and obese (106%), and this was supported by an increase in phospho-ACC in WG. Further, metformin increased IkappaBalpha levels in both WG (150%) and RG (67%) of obese rats, indicative of reduced IKKbeta activity (P < .05), and was associated with reduced IRS1-pSer(307) (30%) in the WG of obese rats (P < .02). From these data we conclude that metformin treatment appears to exert an inhibitory influence on skeletal muscle IKKbeta activity, as evidenced by elevated IkappaBalpha levels and reduced IRS1-Ser(307) phosphorylation in a fiber-type specific manner.
Collapse
Affiliation(s)
- Benjamin T. Bikman
- The Metabolic Institute for the Study of Diabetes and Obesity, East Carolina University, Greenville, NC 27834, USA
- Department of Exercise and Sport Science, East Carolina University, Greenville, NC 27834, USA
- *Benjamin T. Bikman:
| | - Donghai Zheng
- The Metabolic Institute for the Study of Diabetes and Obesity, East Carolina University, Greenville, NC 27834, USA
- Department of Exercise and Sport Science, East Carolina University, Greenville, NC 27834, USA
| | - Daniel A. Kane
- The Metabolic Institute for the Study of Diabetes and Obesity, East Carolina University, Greenville, NC 27834, USA
- Department of Exercise and Sport Science, East Carolina University, Greenville, NC 27834, USA
| | - Ethan J. Anderson
- The Metabolic Institute for the Study of Diabetes and Obesity, East Carolina University, Greenville, NC 27834, USA
- Department of Exercise and Sport Science, East Carolina University, Greenville, NC 27834, USA
| | - Tracey L. Woodlief
- The Metabolic Institute for the Study of Diabetes and Obesity, East Carolina University, Greenville, NC 27834, USA
- Department of Physiology, East Carolina University, Greenville, NC 27834, USA
| | - Jesse W. Price
- Department of Biology, East Carolina University, Greenville, NC 27834, USA
| | - G. Lynis Dohm
- The Metabolic Institute for the Study of Diabetes and Obesity, East Carolina University, Greenville, NC 27834, USA
- Department of Physiology, East Carolina University, Greenville, NC 27834, USA
| | - P. Darrell Neufer
- The Metabolic Institute for the Study of Diabetes and Obesity, East Carolina University, Greenville, NC 27834, USA
- Department of Physiology, East Carolina University, Greenville, NC 27834, USA
| | - Ronald N. Cortright
- The Metabolic Institute for the Study of Diabetes and Obesity, East Carolina University, Greenville, NC 27834, USA
- Department of Physiology, East Carolina University, Greenville, NC 27834, USA
| |
Collapse
|
49
|
Affiliation(s)
- Carsten Schmitz-Peiffer
- Garvan Institute of Medical Research, Sydney, Australia
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Trevor J. Biden
- Garvan Institute of Medical Research, Sydney, Australia
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia
- Corresponding author: Trevor J. Biden,
| |
Collapse
|