1
|
Deane CS, Willis CRG, Gallagher IJ, Brook MS, Gharahdaghi N, Wylie LJ, Wilkinson DJ, Smith K, Atherton PJ, Etheridge T. Nicotinic acid improves mitochondrial function and associated transcriptional pathways in older inactive males. TRANSLATIONAL EXERCISE BIOMEDICINE 2024; 1:277-294. [PMID: 39703532 PMCID: PMC11653476 DOI: 10.1515/teb-2024-0030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 10/30/2024] [Indexed: 12/21/2024]
Abstract
Objectives To examine the effect of the NAD+ precursor, nicotinic acid (NA), for improving skeletal muscle status in sedentary older people. Methods In a double-blind, randomised, placebo-controlled design, 18 sedentary yet otherwise healthy older (65-75 y) males were assigned to 2-weeks of NA (acipimox; 250 mg × 3 daily, n=8) or placebo (PLA, n=10) supplementation. At baseline, and after week 1 and week 2 of supplementation, a battery of functional, metabolic, and molecular readouts were measured. Results Resting and submaximal respiratory exchange ratio was lower (p<0.05) after 2 weeks in the NA group only, but maximal aerobic and anaerobic function and glucose handling were unchanged (p>0.05). Bayesian statistical modelling identified that leak, maximal coupled and maximal uncoupled mitochondrial respiratory states, increased over the 2-week supplemental period in the NA group (probability for a positive change (pd) 85.2, 90.8 and 95.9 %, respectively) but not in PLA. Citrate synthase and protein content of complex II (SDHB) and V (ATP5A) electron transport chain (ETC) components increased over the 2-week period in the NA group only (pd 95.1, 74.5 and 82.3 %, respectively). Mitochondrial and myofibrillar protein synthetic rates remained unchanged in both groups. NA intake altered the muscle transcriptome by increasing the expression of gene pathways related to cell adhesion/cytoskeleton organisation and inflammation/immunity and decreasing pathway expression of ETC and aerobic respiration processes. NAD+-specific pathways (e.g., de novo NAD+ biosynthetic processes) and genes (e.g., NADSYN1) were uniquely regulated by NA. Conclusions NA might be an effective strategy for improving ageing muscle mitochondrial health.
Collapse
Affiliation(s)
- Colleen S. Deane
- Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
- Human Development & Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Craig R. G. Willis
- School of Chemistry and Biosciences, Faculty of Life Sciences, University of Bradford, Bradford, UK
| | - Iain J. Gallagher
- Centre for Biomedicine & Global Health, Edinburgh Napier University, Edinburgh, UK
| | - Matthew S. Brook
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research and National Institute for Health Research Nottingham Biomedical Research Centre, School of Medicine, University of Nottingham, Derby, UK
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Nima Gharahdaghi
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research and National Institute for Health Research Nottingham Biomedical Research Centre, School of Medicine, University of Nottingham, Derby, UK
| | - Lee J. Wylie
- Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Daniel J. Wilkinson
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research and National Institute for Health Research Nottingham Biomedical Research Centre, School of Medicine, University of Nottingham, Derby, UK
| | - Kenneth Smith
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research and National Institute for Health Research Nottingham Biomedical Research Centre, School of Medicine, University of Nottingham, Derby, UK
| | - Philip J. Atherton
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research and National Institute for Health Research Nottingham Biomedical Research Centre, School of Medicine, University of Nottingham, Derby, UK
| | - Timothy Etheridge
- Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| |
Collapse
|
2
|
Feng SS, Wang SJ, Guo L, Ma PP, Ye XL, Pan ML, Hang B, Mao JH, Snijders AM, Lu YB, Ding DF. Serum bile acid and unsaturated fatty acid profiles of non-alcoholic fatty liver disease in type 2 diabetic patients. World J Diabetes 2024; 15:898-913. [PMID: 38766436 PMCID: PMC11099371 DOI: 10.4239/wjd.v15.i5.898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 01/29/2024] [Accepted: 03/14/2024] [Indexed: 05/10/2024] Open
Abstract
BACKGROUND The understanding of bile acid (BA) and unsaturated fatty acid (UFA) profiles, as well as their dysregulation, remains elusive in individuals with type 2 diabetes mellitus (T2DM) coexisting with non-alcoholic fatty liver disease (NAFLD). Investigating these metabolites could offer valuable insights into the pathophy-siology of NAFLD in T2DM. AIM To identify potential metabolite biomarkers capable of distinguishing between NAFLD and T2DM. METHODS A training model was developed involving 399 participants, comprising 113 healthy controls (HCs), 134 individuals with T2DM without NAFLD, and 152 individuals with T2DM and NAFLD. External validation encompassed 172 participants. NAFLD patients were divided based on liver fibrosis scores. The analytical approach employed univariate testing, orthogonal partial least squares-discriminant analysis, logistic regression, receiver operating characteristic curve analysis, and decision curve analysis to pinpoint and assess the diagnostic value of serum biomarkers. RESULTS Compared to HCs, both T2DM and NAFLD groups exhibited diminished levels of specific BAs. In UFAs, particular acids exhibited a positive correlation with NAFLD risk in T2DM, while the ω-6:ω-3 UFA ratio demonstrated a negative correlation. Levels of α-linolenic acid and γ-linolenic acid were linked to significant liver fibrosis in NAFLD. The validation cohort substantiated the predictive efficacy of these biomarkers for assessing NAFLD risk in T2DM patients. CONCLUSION This study underscores the connection between altered BA and UFA profiles and the presence of NAFLD in individuals with T2DM, proposing their potential as biomarkers in the pathogenesis of NAFLD.
Collapse
Affiliation(s)
- Su-Su Feng
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, Jiangsu Province, China
| | - Si-Jing Wang
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, Jiangsu Province, China
| | - Lin Guo
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, Jiangsu Province, China
| | - Pan-Pan Ma
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, Jiangsu Province, China
| | - Xiao-Long Ye
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, Jiangsu Province, China
| | - Ming-Lin Pan
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, Jiangsu Province, China
| | - Bo Hang
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, United States
| | - Jian-Hua Mao
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, United States
| | - Antoine M Snijders
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, United States
| | - Yi-Bing Lu
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, Jiangsu Province, China
| | - Da-Fa Ding
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, Jiangsu Province, China
| |
Collapse
|
3
|
Barssotti L, Soares GM, Marconato-Júnior E, Lourençoni Alves B, Oliveira KM, Carneiro EM, Boschero AC, Barbosa HCL. KSRP improves pancreatic beta cell function and survival. Sci Rep 2024; 14:6136. [PMID: 38480757 PMCID: PMC10937633 DOI: 10.1038/s41598-024-55505-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 02/24/2024] [Indexed: 03/17/2024] Open
Abstract
Impaired insulin production and/or secretion by pancreatic beta cells can lead to high blood glucose levels and type 2 diabetes (T2D). Therefore, investigating new proteins involved in beta cell response to stress conditions could be useful in finding new targets for therapeutic approaches. KH-type splicing regulatory protein (KSRP) is a protein usually involved in gene expression due to its role in post-transcriptional regulation. Although there are studies describing the important role of KSRP in tissues closely related to glucose homeostasis, its effect on pancreatic beta cells has not been explored so far. Pancreatic islets from diet-induced obese mice (C57BL/6JUnib) were used to determine KSRP expression and we also performed in vitro experiments exposing INS-1E cells (pancreatic beta cell line) to different stressors (palmitate or cyclopiazonic acid-CPA) to induce cellular dysfunction. Here we show that KSRP expression is reduced in all the beta cell dysfunction models tested. In addition, when manipulated to knock down KSRP, beta cells exhibited increased death and impaired insulin secretion, whereas KSRP overexpression prevented cell death and increased insulin secretion. Taken together, our findings suggest that KSRP could be an important target to protect beta cells from impaired functioning and death.
Collapse
Affiliation(s)
- Leticia Barssotti
- Obesity and Comorbidities Research Center (OCRC), Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, 13083864, Brazil
| | - Gabriela Moreira Soares
- Obesity and Comorbidities Research Center (OCRC), Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, 13083864, Brazil
| | - Emílio Marconato-Júnior
- Obesity and Comorbidities Research Center (OCRC), Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, 13083864, Brazil
| | - Bruna Lourençoni Alves
- Obesity and Comorbidities Research Center (OCRC), Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, 13083864, Brazil
| | - Kênia Moreno Oliveira
- Obesity and Comorbidities Research Center (OCRC), Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, 13083864, Brazil
| | - Everardo Magalhães Carneiro
- Obesity and Comorbidities Research Center (OCRC), Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, 13083864, Brazil
| | - Antonio Carlos Boschero
- Obesity and Comorbidities Research Center (OCRC), Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, 13083864, Brazil
| | - Helena Cristina Lima Barbosa
- Obesity and Comorbidities Research Center (OCRC), Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, 13083864, Brazil.
| |
Collapse
|
4
|
Rivera Nieves AM, Wauford BM, Fu A. Mitochondrial bioenergetics, metabolism, and beyond in pancreatic β-cells and diabetes. Front Mol Biosci 2024; 11:1354199. [PMID: 38404962 PMCID: PMC10884328 DOI: 10.3389/fmolb.2024.1354199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 01/17/2024] [Indexed: 02/27/2024] Open
Abstract
In Type 1 and Type 2 diabetes, pancreatic β-cell survival and function are impaired. Additional etiologies of diabetes include dysfunction in insulin-sensing hepatic, muscle, and adipose tissues as well as immune cells. An important determinant of metabolic health across these various tissues is mitochondria function and structure. This review focuses on the role of mitochondria in diabetes pathogenesis, with a specific emphasis on pancreatic β-cells. These dynamic organelles are obligate for β-cell survival, function, replication, insulin production, and control over insulin release. Therefore, it is not surprising that mitochondria are severely defective in diabetic contexts. Mitochondrial dysfunction poses challenges to assess in cause-effect studies, prompting us to assemble and deliberate the evidence for mitochondria dysfunction as a cause or consequence of diabetes. Understanding the precise molecular mechanisms underlying mitochondrial dysfunction in diabetes and identifying therapeutic strategies to restore mitochondrial homeostasis and enhance β-cell function are active and expanding areas of research. In summary, this review examines the multidimensional role of mitochondria in diabetes, focusing on pancreatic β-cells and highlighting the significance of mitochondrial metabolism, bioenergetics, calcium, dynamics, and mitophagy in the pathophysiology of diabetes. We describe the effects of diabetes-related gluco/lipotoxic, oxidative and inflammation stress on β-cell mitochondria, as well as the role played by mitochondria on the pathologic outcomes of these stress paradigms. By examining these aspects, we provide updated insights and highlight areas where further research is required for a deeper molecular understanding of the role of mitochondria in β-cells and diabetes.
Collapse
Affiliation(s)
- Alejandra María Rivera Nieves
- Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, United States
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Brian Michael Wauford
- Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, United States
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Accalia Fu
- Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, United States
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, United States
| |
Collapse
|
5
|
Wretlind A, Zobel EH, de Zawadzki A, Ripa RS, Curovic VR, von Scholten BJ, Mattila IM, Hansen TW, Kjær A, Vestergaard H, Rossing P, Legido-Quigley C. Liraglutide Lowers Palmitoleate Levels in Type 2 Diabetes. A Post Hoc Analysis of the LIRAFLAME Randomized Placebo-Controlled Trial. FRONTIERS IN CLINICAL DIABETES AND HEALTHCARE 2022; 3:856485. [PMID: 36992761 PMCID: PMC10012104 DOI: 10.3389/fcdhc.2022.856485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 02/09/2022] [Indexed: 11/13/2022]
Abstract
BackgroundLiraglutide is a glucose-lowering medication used to treat type 2 diabetes and obesity. It is a GLP-1 receptor agonist with downstream metabolic changes beyond the incretin system, such as reducing the risk of cardiovascular complications. The understanding of these changes is critical for improving treatment outcomes. Herein, we present a post hoc experimental analysis using metabolomic phenotyping to discover molecular mecphanisms in response to liraglutide.MethodPlasma samples were obtained from The LiraFlame Study (ClinicalTrials.gov identifier: NCT03449654), a randomized double-blinded placebo-controlled clinical trial, including 102 participants with type 2 diabetes randomized to either liraglutide or placebo treatment for 26 weeks. Mass spectrometry-based metabolomics analyses were carried out on samples from baseline and the end of the trial. Metabolites (n=114) were categorized into pathways and linear mixed models were constructed to evaluate the association between changes in metabolites and liraglutide treatment.ResultsWe found the free fatty acid palmitoleate was significantly reduced in the liraglutide group compared to placebo (adjusted for multiple testing p-value = 0.04). The activity of stearoyl-CoA desaturase-1 (SCD1), the rate limiting enzyme for converting palmitate into palmitoleate, was found significantly downregulated by liraglutide treatment compared to placebo (p-value = 0.01). These metabolic changes have demonstrated to be linked to insulin sensitivity and cardiovascular health.
Collapse
Affiliation(s)
- Asger Wretlind
- Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Rasmus Sejersten Ripa
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | - Andreas Kjær
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Henrik Vestergaard
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Bornholms Hospital, Rønne, Denmark
| | - Peter Rossing
- Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Cristina Legido-Quigley
- Steno Diabetes Center Copenhagen, Herlev, Denmark
- Institute of Pharmaceutical Science, King’s College London, London, United Kingdom
- *Correspondence: Cristina Legido-Quigley,
| |
Collapse
|
6
|
Vilas-Boas EA, Almeida DC, Roma LP, Ortis F, Carpinelli AR. Lipotoxicity and β-Cell Failure in Type 2 Diabetes: Oxidative Stress Linked to NADPH Oxidase and ER Stress. Cells 2021; 10:cells10123328. [PMID: 34943836 PMCID: PMC8699655 DOI: 10.3390/cells10123328] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/18/2021] [Accepted: 11/22/2021] [Indexed: 12/17/2022] Open
Abstract
A high caloric intake, rich in saturated fats, greatly contributes to the development of obesity, which is the leading risk factor for type 2 diabetes (T2D). A persistent caloric surplus increases plasma levels of fatty acids (FAs), especially saturated ones, which were shown to negatively impact pancreatic β-cell function and survival in a process called lipotoxicity. Lipotoxicity in β-cells activates different stress pathways, culminating in β-cells dysfunction and death. Among all stresses, endoplasmic reticulum (ER) stress and oxidative stress have been shown to be strongly correlated. One main source of oxidative stress in pancreatic β-cells appears to be the reactive oxygen species producer NADPH oxidase (NOX) enzyme, which has a role in the glucose-stimulated insulin secretion and in the β-cell demise during both T1 and T2D. In this review, we focus on the acute and chronic effects of FAs and the lipotoxicity-induced β-cell failure during T2D development, with special emphasis on the oxidative stress induced by NOX, the ER stress, and the crosstalk between NOX and ER stress.
Collapse
Affiliation(s)
- Eloisa Aparecida Vilas-Boas
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo 05508-000, Brazil
- Department of Biochemistry, Institute of Chemistry, University of São Paulo (USP), São Paulo 05508-900, Brazil
- Correspondence: (E.A.V.-B.); (A.R.C.); Tel.: +55-(11)-3091-7246 (A.R.C.)
| | - Davidson Correa Almeida
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo 05508-000, Brazil; (D.C.A.); (F.O.)
| | - Leticia Prates Roma
- Center for Human and Molecular Biology (ZHMB), Department of Biophysics, Saarland University, 66424 Homburg, Germany;
| | - Fernanda Ortis
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo 05508-000, Brazil; (D.C.A.); (F.O.)
| | - Angelo Rafael Carpinelli
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo 05508-000, Brazil
- Correspondence: (E.A.V.-B.); (A.R.C.); Tel.: +55-(11)-3091-7246 (A.R.C.)
| |
Collapse
|
7
|
Koduru SV, Leberfinger AN, Ozbolat IT, Ravnic DJ. Navigating the Genomic Landscape of Human Adipose Stem Cell-Derived β-Cells. Stem Cells Dev 2021; 30:1153-1170. [PMID: 34514867 DOI: 10.1089/scd.2021.0160] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Diabetes is a pandemic manifested through glucose dysregulation mediated by inadequate insulin secretion by beta cells. A beta cell replacement strategy would transform the treatment paradigm from pharmacologic glucose modulation to a genuine cure. Stem cells have emerged as a potential source for beta cell (β-cell) engineering. The detailed generation of functional β-cells from both embryonic and induced pluripotent stem cells has recently been described. Adult stem cells, including adipose derived, may also offer a therapeutic approach, but remain ill defined. In our study, we performed an in-depth assessment of insulin-producing beta cells generated from human adipose, irrespective of donor patient age, gender, and health status. Cellular transformation was confirmed using flow cytometry and single-cell imaging. Insulin secretion was observed with glucose stimulation and abrogated following palmitate exposure, a common free fatty acid implicated in human beta cell dysfunction. We used next-generation sequencing to explore gene expression changes before and after differentiation of patient-matched samples, which revealed more than 5,000 genes enriched. Adipose-derived beta cells displayed comparable gene expression to native β-cells. Pathway analysis demonstrated relevance to stem cell differentiation and pancreatic developmental processes, which are vital to cellular function, structural development, and regulation. We conclude that the functions associated with adipose derived beta cells are mediated through relevant changes in the transcriptome, which resemble those seen in native β-cell morphogenesis and maturation. Therefore, they may represent a viable option for the clinical translation of stem cell-based therapies in diabetes.
Collapse
Affiliation(s)
- Srinivas V Koduru
- Irvin S. Zubar Plastic Surgery Research Laboratory, Penn State College of Medicine, Hershey, Pennsylvania, USA.,Division of Plastic Surgery, Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA.,Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Ashley N Leberfinger
- Irvin S. Zubar Plastic Surgery Research Laboratory, Penn State College of Medicine, Hershey, Pennsylvania, USA.,Division of Plastic Surgery, Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Ibrahim T Ozbolat
- Department of Biomedical Engineering, Materials Research Institute, The Huck Institutes of Life Sciences, Penn State University, University Park, Pennsylvania, USA.,Engineering Science and Mechanics Department, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Dino J Ravnic
- Irvin S. Zubar Plastic Surgery Research Laboratory, Penn State College of Medicine, Hershey, Pennsylvania, USA.,Division of Plastic Surgery, Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| |
Collapse
|
8
|
Lipotoxic Impairment of Mitochondrial Function in β-Cells: A Review. Antioxidants (Basel) 2021; 10:antiox10020293. [PMID: 33672062 PMCID: PMC7919463 DOI: 10.3390/antiox10020293] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 02/06/2021] [Accepted: 02/11/2021] [Indexed: 02/08/2023] Open
Abstract
Lipotoxicity is a major contributor to type 2 diabetes mainly promoting mitochondrial dysfunction. Lipotoxic stress is mediated by elevated levels of free fatty acids through various mechanisms and pathways. Impaired peroxisome proliferator-activated receptor (PPAR) signaling, enhanced oxidative stress levels, and uncoupling of the respiratory chain result in ATP deficiency, while β-cell viability can be severely impaired by lipotoxic modulation of PI3K/Akt and mitogen-activated protein kinase (MAPK)/extracellular-signal-regulated kinase (ERK) pathways. However, fatty acids are physiologically required for an unimpaired β-cell function. Thus, preparation, concentration, and treatment duration determine whether the outcome is beneficial or detrimental when fatty acids are employed in experimental setups. Further, ageing is a crucial contributor to β-cell decay. Cellular senescence is connected to loss of function in β-cells and can further be promoted by lipotoxicity. The potential benefit of nutrients has been broadly investigated, and particularly polyphenols were shown to be protective against both lipotoxicity and cellular senescence, maintaining the physiology of β-cells. Positive effects on blood glucose regulation, mitigation of oxidative stress by radical scavenging properties or regulation of antioxidative enzymes, and modulation of apoptotic factors were reported. This review summarizes the significance of lipotoxicity and cellular senescence for mitochondrial dysfunction in the pancreatic β-cell and outlines potential beneficial effects of plant-based nutrients by the example of polyphenols.
Collapse
|
9
|
Vilas-Boas EA, Nalbach L, Ampofo E, Lucena CF, Naudet L, Ortis F, Carpinelli AR, Morgan B, Roma LP. Transient NADPH oxidase 2-dependent H 2O 2 production drives early palmitate-induced lipotoxicity in pancreatic islets. Free Radic Biol Med 2021; 162:1-13. [PMID: 33249137 DOI: 10.1016/j.freeradbiomed.2020.11.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/18/2020] [Accepted: 11/20/2020] [Indexed: 01/12/2023]
Abstract
Modern lifestyles, including lack of physical activity and poor nutritional habits, are driving the rapidly increasing prevalence of obesity and type 2 diabetes. Increased levels of free fatty acids (FFAs), particularly saturated FFAs, in obese individuals have been linked to pancreatic β-cell failure. This process, termed lipotoxicity, involves activation of several stress responses, including ER stress and oxidative stress. However, the molecular underpinnings and causal relationships between the disparate stress responses remain unclear. Here we employed transgenic mice, expressing a genetically-encoded cytosolic H2O2 sensor, roGFP2-Orp1, to monitor dynamic changes in H2O2 levels in pancreatic islets in response to chronic palmitate exposure. We identified a transient increase in H2O2 levels from 4 to 8 h after palmitate addition, which was mirrored by a concomitant decrease in cellular NAD(P)H levels. Intriguingly, islets isolated from NOX2 knock-out mice displayed no H2O2 transient upon chronic palmitate treatment. Furthermore, NOX2 knockout rescued palmitate-dependent impairment of insulin secretion, calcium homeostasis and viability. Chemical inhibition of NOX activity protected islets from palmitate-induced impairment in insulin secretion, however had no detectable impact upon the induction of ER stress. In summary, our results reveal that transient NOX2-dependent H2O2 production is a likely cause of early palmitate-dependent lipotoxic effects.
Collapse
Affiliation(s)
- Eloisa Aparecida Vilas-Boas
- Department of Biophysics, Center for Human and Molecular Biology (ZHMB), Saarland University, Homburg, Germany; Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Lisa Nalbach
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - Emmanuel Ampofo
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - Camila Ferraz Lucena
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Léa Naudet
- Department of Biophysics, Center for Human and Molecular Biology (ZHMB), Saarland University, Homburg, Germany
| | - Fernanda Ortis
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Angelo Rafael Carpinelli
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Bruce Morgan
- Institute for Biochemistry, Center for Human and Molecular Biology (ZHMB), Saarland University, Saarbrücken, Germany
| | - Leticia Prates Roma
- Department of Biophysics, Center for Human and Molecular Biology (ZHMB), Saarland University, Homburg, Germany.
| |
Collapse
|
10
|
Callebaut A, Derua R, Vig S, Delong T, Mathieu C, Overbergh L. Identification of Deamidated Peptides in Cytokine-Exposed MIN6 Cells through LC-MS/MS Using a Shortened Digestion Time and Inspection of MS2 Spectra. J Proteome Res 2020; 20:1405-1414. [PMID: 33372785 DOI: 10.1021/acs.jproteome.0c00801] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Enzymatic deamidation, the conversion of glutamine (Gln) into glutamic acid (Glu) residues, mediated by tissue transglutaminase enzymes, can provoke autoimmunity by generating altered self-epitopes, a process well-known in celiac disease and more recently also described in type 1 diabetes (T1D). To identify deamidated proteins, liquid chromatography-tandem mass spectrometry is the method of choice. However, as nonenzymatic deamidations on asparagine (Asn) and to a minor extent on Gln are frequently induced in vitro during proteomics sample preparation, the accurate detection of in vivo deamidation can be hampered. Here we report on the optimization of a method to reduce in vitro generated deamidation by 70% using improved trypsin digestion conditions (90 min/pH 8). We also point to the critical importance of manual inspection of MS2 spectra, considering that only 55% of the high quality peptides with Gln deamidation were assigned correctly using an automated search algorithm. As proof of principal, using these criteria, we showed a significant increase in levels of both Asn and Gln deamidation in cytokine-exposed murine MIN6 β-cells, paralleled by an increase in tissue transglutaminase activity. These findings add evidence to the hypothesis that deamidation is occurring in stressed β-cell proteins and can be involved in the autoimmune process in T1D.
Collapse
Affiliation(s)
- Aïsha Callebaut
- Laboratory for Clinical and Experimental Endocrinology, KU Leuven, 3000 Leuven, Belgium
| | - Rita Derua
- Laboratory of Protein Phosphorylation and Proteomics, KU Leuven, 3000 Leuven, Belgium.,SyBioMa, KU Leuven, 3000 Leuven, Belgium
| | - Saurabh Vig
- Laboratory for Clinical and Experimental Endocrinology, KU Leuven, 3000 Leuven, Belgium
| | - Thomas Delong
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz, Aurora, Colorado 80045, United States
| | - Chantal Mathieu
- Laboratory for Clinical and Experimental Endocrinology, KU Leuven, 3000 Leuven, Belgium
| | - Lut Overbergh
- Laboratory for Clinical and Experimental Endocrinology, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
11
|
Pu ZQ, Liu D, Lobo Mouguegue HPP, Jin CW, Sadiq E, Qin DD, Yu TF, Zong C, Chen JC, Zhao RX, Lin JY, Cheng J, Yu X, Li X, Zhang YC, Liu YT, Guan QB, Wang XD. NR4A1 counteracts JNK activation incurred by ER stress or ROS in pancreatic β-cells for protection. J Cell Mol Med 2020; 24:14171-14183. [PMID: 33124187 PMCID: PMC7754045 DOI: 10.1111/jcmm.16028] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/08/2020] [Accepted: 10/05/2020] [Indexed: 12/20/2022] Open
Abstract
Sustained hyperglycaemia and hyperlipidaemia incur endoplasmic reticulum stress (ER stress) and reactive oxygen species (ROS) overproduction in pancreatic β‐cells. ER stress or ROS causes c‐Jun N‐terminal kinase (JNK) activation, and the activated JNK triggers apoptosis in different cells. Nuclear receptor subfamily 4 group A member 1 (NR4A1) is an inducible multi‐stress response factor. The aim of this study was to explore the role of NR4A1 in counteracting JNK activation induced by ER stress or ROS and the related mechanism. qPCR, Western blotting, dual‐luciferase reporter and ChIP assays were applied to detect gene expression or regulation by NR4A1. Immunofluorescence was used to detect a specific protein expression in β‐cells. Our data showed that NR4A1 reduced the phosphorylated JNK (p‐JNK) in MIN6 cells encountering ER stress or ROS and reduced MKK4 protein in a proteasome‐dependent manner. We found that NR4A1 increased the expression of cbl‐b (an E3 ligase); knocking down cbl‐b expression increased MKK4 and p‐JNK levels under ER stress or ROS conditions. We elucidated that NR4A1 enhanced the transactivation of cbl‐b promoter by physical association. We further confirmed that cbl‐b expression in β‐cells was reduced in NR4A1‐knockout mice compared with WT mice. NR4A1 down‐regulates JNK activation by ER stress or ROS in β‐cells via enhancing cbl‐b expression.
Collapse
Affiliation(s)
- Ze-Qing Pu
- Department of Cell Biology, Shandong University School of Medicine, Jinan, China
| | - Dong Liu
- Department of Cell Biology, Shandong University School of Medicine, Jinan, China
| | | | - Cheng-Wen Jin
- Department of Cell Biology, Shandong University School of Medicine, Jinan, China
| | - Esha Sadiq
- Department of Cell Biology, Shandong University School of Medicine, Jinan, China
| | - Dan-Dan Qin
- Department of Cell Biology, Shandong University School of Medicine, Jinan, China
| | - Tian-Fu Yu
- Department of Cell Biology, Shandong University School of Medicine, Jinan, China
| | - Chen Zong
- Department of Cell Biology, Shandong University School of Medicine, Jinan, China
| | - Ji-Cui Chen
- Blood Transfusion Department, Qilu Hospital of Shandong University, Jinan, China
| | - Ru-Xing Zhao
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, China
| | - Jing-Yu Lin
- Department of Physiology, Shandong University School of Medicine, Jinan, China
| | - Jie Cheng
- Department of Physiology, Shandong University School of Medicine, Jinan, China
| | - Xiao Yu
- Department of Physiology, Shandong University School of Medicine, Jinan, China.,Key Laboratory of Protein Sciences for Chronic Degenerative Diseases in Universities of Shandong (Shandong University), Jinan, China
| | - Xia Li
- Department of Cell Biology, Shandong University School of Medicine, Jinan, China
| | - Yu-Chao Zhang
- Department of Endocrinology, Qingdao Municipal Hospital, Qingdao, China
| | - Yuan-Tao Liu
- Department of Endocrinology, Qingdao Municipal Hospital, Qingdao, China
| | - Qing-Bo Guan
- Department of Endocrinology, Shandong Provincial Hospital, Affiliated to Shandong University, Jinan, China
| | - Xiang-Dong Wang
- Department of Cell Biology, Shandong University School of Medicine, Jinan, China.,Key Laboratory of Protein Sciences for Chronic Degenerative Diseases in Universities of Shandong (Shandong University), Jinan, China
| |
Collapse
|
12
|
Lytrivi M, Ghaddar K, Lopes M, Rosengren V, Piron A, Yi X, Johansson H, Lehtiö J, Igoillo-Esteve M, Cunha DA, Marselli L, Marchetti P, Ortsäter H, Eizirik DL, Cnop M. Combined transcriptome and proteome profiling of the pancreatic β-cell response to palmitate unveils key pathways of β-cell lipotoxicity. BMC Genomics 2020; 21:590. [PMID: 32847508 PMCID: PMC7448506 DOI: 10.1186/s12864-020-07003-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 08/19/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Prolonged exposure to elevated free fatty acids induces β-cell failure (lipotoxicity) and contributes to the pathogenesis of type 2 diabetes. In vitro exposure of β-cells to the saturated free fatty acid palmitate is a valuable model of lipotoxicity, reproducing features of β-cell failure observed in type 2 diabetes. In order to map the β-cell response to lipotoxicity, we combined RNA-sequencing of palmitate-treated human islets with iTRAQ proteomics of insulin-secreting INS-1E cells following a time course exposure to palmitate. RESULTS Crossing transcriptome and proteome of palmitate-treated β-cells revealed 85 upregulated and 122 downregulated genes at both transcript and protein level. Pathway analysis identified lipid metabolism, oxidative stress, amino-acid metabolism and cell cycle pathways among the most enriched palmitate-modified pathways. Palmitate induced gene expression changes compatible with increased free fatty acid mitochondrial import and β-oxidation, decreased lipogenesis and modified cholesterol transport. Palmitate modified genes regulating endoplasmic reticulum (ER) function, ER-to-Golgi transport and ER stress pathways. Furthermore, palmitate modulated cAMP/protein kinase A (PKA) signaling, inhibiting expression of PKA anchoring proteins and downregulating the GLP-1 receptor. SLC7 family amino-acid transporters were upregulated in response to palmitate but this induction did not contribute to β-cell demise. To unravel critical mediators of lipotoxicity upstream of the palmitate-modified genes, we identified overrepresented transcription factor binding sites and performed network inference analysis. These identified LXR, PPARα, FOXO1 and BACH1 as key transcription factors orchestrating the metabolic and oxidative stress responses to palmitate. CONCLUSIONS This is the first study to combine transcriptomic and sensitive time course proteomic profiling of palmitate-exposed β-cells. Our results provide comprehensive insight into gene and protein expression changes, corroborating and expanding beyond previous findings. The identification of critical drivers and pathways of the β-cell lipotoxic response points to novel therapeutic targets for type 2 diabetes.
Collapse
Affiliation(s)
- Maria Lytrivi
- ULB Center for Diabetes Research, Université Libre de Bruxelles, CP-618, Route de Lennik 808, 1070, Brussels, Belgium.,Division of Endocrinology, Erasmus Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Kassem Ghaddar
- ULB Center for Diabetes Research, Université Libre de Bruxelles, CP-618, Route de Lennik 808, 1070, Brussels, Belgium
| | - Miguel Lopes
- ULB Center for Diabetes Research, Université Libre de Bruxelles, CP-618, Route de Lennik 808, 1070, Brussels, Belgium
| | - Victoria Rosengren
- Diabetes Research Unit, Department of Clinical Science and Education, Sodersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Anthony Piron
- ULB Center for Diabetes Research, Université Libre de Bruxelles, CP-618, Route de Lennik 808, 1070, Brussels, Belgium
| | - Xiaoyan Yi
- ULB Center for Diabetes Research, Université Libre de Bruxelles, CP-618, Route de Lennik 808, 1070, Brussels, Belgium
| | - Henrik Johansson
- Clinical Proteomics Mass Spectrometry, Department of Oncology-Pathology, Karolinska Institutet, Science for Life Laboratory, 171 21, Solna, Sweden
| | - Janne Lehtiö
- Clinical Proteomics Mass Spectrometry, Department of Oncology-Pathology, Karolinska Institutet, Science for Life Laboratory, 171 21, Solna, Sweden
| | - Mariana Igoillo-Esteve
- ULB Center for Diabetes Research, Université Libre de Bruxelles, CP-618, Route de Lennik 808, 1070, Brussels, Belgium
| | - Daniel A Cunha
- ULB Center for Diabetes Research, Université Libre de Bruxelles, CP-618, Route de Lennik 808, 1070, Brussels, Belgium
| | - Lorella Marselli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Henrik Ortsäter
- Diabetes Research Unit, Department of Clinical Science and Education, Sodersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Decio L Eizirik
- ULB Center for Diabetes Research, Université Libre de Bruxelles, CP-618, Route de Lennik 808, 1070, Brussels, Belgium
| | - Miriam Cnop
- ULB Center for Diabetes Research, Université Libre de Bruxelles, CP-618, Route de Lennik 808, 1070, Brussels, Belgium. .,Division of Endocrinology, Erasmus Hospital, Université Libre de Bruxelles, Brussels, Belgium.
| |
Collapse
|
13
|
Jin MH, Shen GN, Jin YH, Sun HN, Zhen X, Zhang YQ, Lee DS, Cui YD, Yu LY, Kim JS, Kwon T, Han YH. Peroxiredoxin I deficiency increases pancreatic β‑cell apoptosis after streptozotocin stimulation via the AKT/GSK3β signaling pathway. Mol Med Rep 2020; 22:1831-1838. [PMID: 32705184 PMCID: PMC7411341 DOI: 10.3892/mmr.2020.11279] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 05/28/2020] [Indexed: 01/11/2023] Open
Abstract
Apoptosis of pancreatic β-cells is involved in the pathogenesis of type I and II diabetes. Peroxiredoxin I (Prx I) serves an important role in regulating cellular apoptosis; however, the role of Prx I in pancreatic β-cell apoptosis is not completely understood. In the present study, the role of peroxiredoxin 1 (Prx I) during streptozotocin (STZ)-induced apoptosis of pancreatic β-cells was investigated. The expression level of Prx I was decreased by STZ treatment in a time-dependent manner, and apoptosis of Prx I knockdown MIN6 cells was increased by STZ stimulation, compared with untransduced MIN6 cells. Furthermore, an intraperitoneal injection of STZ increased pancreatic islet damage in Prx I knockout mice, compared with wild-type and Prx II knockout mice. AKT and glycogen synthase kinase (GSK)-3β phosphorylation significantly decreased following Prx I knockdown in MIN6 cells. However, phosphorylated β-catenin and p65 levels significantly increased after STZ stimulation, compared with untransduced cells. The results of the present study indicate that deletion of Prx I mediated STZ-induced pancreatic β-cell death in vivo and in vitro by regulating the AKT/GSK-3β/β-catenin signaling pathway, as well as NF-κB signaling. These findings provide a theoretical basis for treatment of pancreatic damage.
Collapse
Affiliation(s)
- Mei-Hua Jin
- Laboratory of Disease Model Research Center, College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Gui-Nan Shen
- Laboratory of Disease Model Research Center, College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Ying-Hua Jin
- Department of Library and Information Center, Library of Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Hu-Nan Sun
- Laboratory of Disease Model Research Center, College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Xing Zhen
- Laboratory of Disease Model Research Center, College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Yong-Qing Zhang
- Laboratory of Disease Model Research Center, College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Dong-Seok Lee
- School of Life Sciences, KUN Creative Bioresearch Group, Kyungpook National University, Daegu, Gyeongsangbuk 702‑701, Republic of Korea
| | - Yu-Dong Cui
- Laboratory of Disease Model Research Center, College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Li-Yun Yu
- Laboratory of Disease Model Research Center, College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Ji-Su Kim
- Primate Resources Center, Korea Research Institute of Bioscience and Biotechnology, Ibam‑myeon, Jeongeup‑si, Jeonbuk 56216, Republic of Korea
| | - Taeho Kwon
- Primate Resources Center, Korea Research Institute of Bioscience and Biotechnology, Ibam‑myeon, Jeongeup‑si, Jeonbuk 56216, Republic of Korea
| | - Ying-Hao Han
- Laboratory of Disease Model Research Center, College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| |
Collapse
|
14
|
Vilas-Boas EA, Karabacz N, Marsiglio-Librais GN, Valle MMR, Nalbach L, Ampofo E, Morgan B, Carpinelli AR, Roma LP. Chronic activation of GPR40 does not negatively impact upon BRIN-BD11 pancreatic β-cell physiology and function. Pharmacol Rep 2020; 72:1725-1737. [PMID: 32274767 PMCID: PMC7704488 DOI: 10.1007/s43440-020-00101-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/18/2020] [Accepted: 03/21/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Free fatty acids (FFAs) are known for their dual effects on insulin secretion and pancreatic β-cell survival. Short-term exposure to FFAs, such as palmitate, increases insulin secretion. On the contrary, long-term exposure to saturated FFAs results in decreased insulin secretion, as well as triggering oxidative stress and endoplasmic reticulum (ER) stress, culminating in cell death. The effects of FFAs can be mediated either via their intracellular oxidation and consequent effects on cellular metabolism or via activation of the membrane receptor GPR40. Both pathways are likely to be activated upon both short- and long-term exposure to FFAs. However, the precise role of GPR40 in β-cell physiology, especially upon chronic exposure to FFAs, remains unclear. METHODS We used the GPR40 agonist (GW9508) and antagonist (GW1100) to investigate the impact of chronically modulating GPR40 activity on BRIN-BD11 pancreatic β-cells physiology and function. RESULTS We observed that chronic activation of GPR40 did not lead to increased apoptosis, and both proliferation and glucose-induced calcium entry were unchanged compared to control conditions. We also observed no increase in H2O2 or superoxide levels and no increase in the ER stress markers p-eIF2α, CHOP and BIP. As expected, palmitate led to increased H2O2 levels, decreased cell viability and proliferation, as well as decreased metabolism and calcium entry. These changes were not counteracted by the co-treatment of palmitate-exposed cells with the GPR40 antagonist GW1100. CONCLUSIONS Chronic activation of GPR40 using GW9508 does not negatively impact upon BRIN-BD11 pancreatic β-cells physiology and function. The GPR40 antagonist GW1100 does not protect against the deleterious effects of chronic palmitate exposure. We conclude that GPR40 is probably not involved in mediating the toxicity associated with chronic palmitate exposure.
Collapse
Affiliation(s)
- Eloisa Aparecida Vilas-Boas
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo (USP), Sao Paulo, SP, Brazil.,Department of Biophysics, Center for Human and Molecular Biology, Saarland University, Universität Des Saarlandes, CIPMM, Geb. 48, 66421, Homburg/Saar, Germany
| | - Noémie Karabacz
- Department of Biophysics, Center for Human and Molecular Biology, Saarland University, Universität Des Saarlandes, CIPMM, Geb. 48, 66421, Homburg/Saar, Germany
| | | | - Maíra Melo Rezende Valle
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - Lisa Nalbach
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg/Saar, Germany
| | - Emmanuel Ampofo
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg/Saar, Germany
| | - Bruce Morgan
- Institute of Biochemistry, Center for Human and Molecular Biology (ZHMB), Saarland University, 66123, Saarbrücken, Germany
| | - Angelo Rafael Carpinelli
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - Leticia Prates Roma
- Department of Biophysics, Center for Human and Molecular Biology, Saarland University, Universität Des Saarlandes, CIPMM, Geb. 48, 66421, Homburg/Saar, Germany.
| |
Collapse
|
15
|
Nrf2 Activation Protects Mouse Beta Cells from Glucolipotoxicity by Restoring Mitochondrial Function and Physiological Redox Balance. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:7518510. [PMID: 31827698 PMCID: PMC6885177 DOI: 10.1155/2019/7518510] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/31/2019] [Accepted: 09/06/2019] [Indexed: 12/25/2022]
Abstract
Influencing the redox balance of pancreatic beta cells could be a promising strategy for the treatment of diabetes. Nuclear factor erythroid 2p45-related factor 2 (Nrf2) is present in beta cells and regulates numerous genes involved in antioxidant defense. As reactive oxygen species (ROS) are important for beta cell signaling but induce oxidative stress when present in excess, this study elucidates the influence of Nrf2-activating compounds on different kinds of ROS and correlates changes in redox balance to effects on mitochondrial function, insulin release, and cell viability. Acute glucose stimulation (15 mmol/L) of murine islet cells of C57Bl/6N mice affects ROS and redox status of the cells differently. Those ROS monitored by dihydroethidium, which detects superoxide radical anions, decrease. By contrast, oxidant status, monitored by dichlorodihydrofluorescein, as well as intracellular H2O2, increases. Glucolipotoxicity completely prevents these fast, glucose-mediated alterations and inhibits glucose-induced NAD(P)H production, mitochondrial hyperpolarization, and ATP synthesis. Oltipraz (10 μmol/L) or dimethyl fumarate (DMF, 50 μmol/L) leads to nuclear accumulation of Nrf2, restores mitochondrial activity and glucose-dependent ROS turnover, and antagonizes glucolipotoxicity-induced inhibition of insulin release and apoptosis. Importantly, these beneficial effects only occur when beta cells are challenged and damaged by high lipid and carbohydrate supply. At physiological conditions, insulin release is markedly reduced in response to both Nrf2 activators. This is not associated with severe impairment of glucose-induced mitochondrial hyperpolarization or a rise in apoptosis but coincides with altered ROS handling. In conclusion, Nrf2 activators protect beta cells against glucolipotoxicity by preserving mitochondrial function and redox balance. As our data show that this maintains glucose-stimulated insulin secretion, targeting Nrf2 might be suited to ameliorate progression of type 2 diabetes mellitus. By contrast, nonstressed beta cells do not benefit from Nrf2 activation, thus underlining the importance of physiological shifts in ROS homeostasis for the regulation of beta cell function.
Collapse
|
16
|
Li Z, Zhou M, Cai Z, Liu H, Zhong W, Hao Q, Cheng D, Hu X, Hou J, Xu P, Xue Y, Zhou Y, Xu T. RNA-binding protein DDX1 is responsible for fatty acid-mediated repression of insulin translation. Nucleic Acids Res 2019; 46:12052-12066. [PMID: 30295850 PMCID: PMC6294501 DOI: 10.1093/nar/gky867] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Accepted: 09/14/2018] [Indexed: 01/13/2023] Open
Abstract
The molecular mechanism in pancreatic β cells underlying hyperlipidemia and insulin insufficiency remains unclear. Here, we find that the fatty acid-induced decrease in insulin levels occurs due to a decrease in insulin translation. Since regulation at the translational level is generally mediated through RNA-binding proteins, using RNA antisense purification coupled with mass spectrometry, we identify a novel insulin mRNA-binding protein, namely, DDX1, that is sensitive to palmitate treatment. Notably, the knockdown or overexpression of DDX1 affects insulin translation, and the knockdown of DDX1 eliminates the palmitate-induced repression of insulin translation. Molecular mechanism studies show that palmitate treatment causes DDX1 phosphorylation at S295 and dissociates DDX1 from insulin mRNA, thereby leading to the suppression of insulin translation. In addition, DDX1 may interact with the translation initiation factors eIF3A and eIF4B to regulate translation. In high-fat diet mice, the inhibition of insulin translation happens at an early prediabetic stage before the elevation of glucose levels. We speculate that the DDX1-mediated repression of insulin translation worsens the situation of insulin resistance and contributes to the elevation of blood glucose levels in obese animals.
Collapse
Affiliation(s)
- Zonghong Li
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics & University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China.,Jilin Province Key Laboratory on Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China
| | - Maoge Zhou
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics & University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhaokui Cai
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Hongyang Liu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics & University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Wen Zhong
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics & University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Qiang Hao
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics & University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Dongwan Cheng
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics & University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Xihao Hu
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Junjie Hou
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics & University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Pingyong Xu
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yuanchao Xue
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yifa Zhou
- Jilin Province Key Laboratory on Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China
| | - Tao Xu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics & University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
17
|
Nie Y, Li J, Jin Y, Nyomba BLG, Cattini PA, Vakili H. Negative Effects of Cyclic Palmitate Treatment on Glucose Responsiveness and Insulin Production in Mouse Insulinoma Min6 Cells Are Reversible. DNA Cell Biol 2019; 38:395-403. [PMID: 30702352 DOI: 10.1089/dna.2018.4558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Pancreatic β-cell failure is characterized by compromised insulin secretion in response to glucose, which ultimately results in hyperglycemia, the clinical hallmark of type 2 diabetes mellitus (T2DM). Acute exposure to plasma free fatty acids (FFAs) potentiates glucose stimulated insulin secretion (GSIS), while chronic exposure impairs GSIS, and the latter has been associated with the mechanism of β cell failure in obesity linked T2DM. By contrast, growth hormone (GH) signaling has been linked positively to GSIS in β cells. Numerous studies have examined chronic exposure of β cells to elevated FFAs both with in vivo cohorts and in vitro models. Little attention, however, has been given to the fluctuation of plasma FFA levels due to rhythmic effects that are affected by daily diet and fat intake. Mouse insulinoma Min6 cells were exposed to cyclic/daily palmitate treatment over 2 and 3 days to assess effects on GSIS. Cyclic/daily palmitate treatment with a period of recovery negatively affected GSIS in a dose-dependent manner. Removal of palmitate after two cycles/day resulted in reversal of the effect on GSIS, which was also reflected by relative gene expression involved in insulin biosynthesis (Ins1, Ins2, Pdx1, and MafA) and GSIS (glucose 2 transporter and glucokinase). Modest positive effects on GSIS and glucokinase transcript levels were also observed when Min6 cells were cotreated with human GH and palmitate. These observations indicate that like continuous palmitate treatment, cyclic exposure to palmitate can acutely impair GSIS over 48 and 72 h. However, they also suggest that the negative effects of short periods of exposure to FFAs on β cell function remain reversible.
Collapse
Affiliation(s)
- Yuanyuan Nie
- 1 Stem Cell and Cancer Center, Jilin University, Changchun, Jilin, China
| | - Jiaxuan Li
- 1 Stem Cell and Cancer Center, Jilin University, Changchun, Jilin, China
| | - Yan Jin
- 2 Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - B L Grégoire Nyomba
- 3 Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Peter A Cattini
- 2 Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Hana Vakili
- 4 Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
18
|
Němcová-Fürstová V, Balušíková K, Halada P, Pavlíková N, Šrámek J, Kovář J. Stearate-Induced Apoptosis in Human Pancreatic β-Cells is Associated with Changes in Membrane Protein Expression and These Changes are Inhibited by Oleate. Proteomics Clin Appl 2019; 13:e1800104. [PMID: 30666801 DOI: 10.1002/prca.201800104] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 12/12/2018] [Indexed: 12/25/2022]
Abstract
PURPOSE Lipotoxicity is implicated in type 2 diabetes pathogenesis. Its molecular mechanisms are not completely understood. The aim of this study is to identify new suspect proteins involved in pancreatic β-cell death induction by saturated fatty acids and its inhibition by unsaturated fatty acids. EXPERIMENTAL DESIGN Employing 2DE analysis and subsequent western blot confirmation, the differences in membrane/membrane-associated protein expression in human β-cell line NES2Y are assessed during cell death induction by stearate and its inhibition by oleate. RESULTS Induction of apoptosis by stearate is associated with significantly increased levels of Hsp90β, peroxiredoxin-1, and 14-3-3γ in the membrane fraction of NES2Y cells and significantly decreased levels of annexin A2, annexin A4, and reticulocalbin-2. All these changes are significantly inhibited by oleate co-application. No expression changes are detected after application of stearate together with oleate. Furthermore, the expression of reticulocalbin-2 is significantly decreased after stearate application also in the whole cell lysate. CONCLUSIONS AND CLINICAL RELEVANCE Several membrane-associated proteins that could be related to pro- and anti-apoptotic signaling initiated by fatty acids in human pancreatic β-cells are identified. As far as we know, annexin A4, reticulocalbin-2, and 14-3-3γ represent novel molecules related to the effect of fatty acids on β-cell viability.
Collapse
Affiliation(s)
- Vlasta Němcová-Fürstová
- Department of Biochemistry, Cell and Molecular Biology & Center for Research of Diabetes, Metabolism and Nutrition, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Kamila Balušíková
- Department of Biochemistry, Cell and Molecular Biology & Center for Research of Diabetes, Metabolism and Nutrition, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Petr Halada
- Laboratory of Molecular Structure Characterization, Institute of Microbiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Nela Pavlíková
- Department of Biochemistry, Cell and Molecular Biology & Center for Research of Diabetes, Metabolism and Nutrition, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jan Šrámek
- Department of Biochemistry, Cell and Molecular Biology & Center for Research of Diabetes, Metabolism and Nutrition, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jan Kovář
- Department of Biochemistry, Cell and Molecular Biology & Center for Research of Diabetes, Metabolism and Nutrition, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
19
|
Bugliani M, Mossuto S, Grano F, Suleiman M, Marselli L, Boggi U, De Simone P, Eizirik DL, Cnop M, Marchetti P, De Tata V. Modulation of Autophagy Influences the Function and Survival of Human Pancreatic Beta Cells Under Endoplasmic Reticulum Stress Conditions and in Type 2 Diabetes. Front Endocrinol (Lausanne) 2019; 10:52. [PMID: 30863363 PMCID: PMC6399112 DOI: 10.3389/fendo.2019.00052] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 01/21/2019] [Indexed: 01/09/2023] Open
Abstract
Autophagy is the major mechanism involved in degradation and recycling of intracellular components, and its alterations have been proposed to cause beta cell dysfunction. In this study, we explored the effects of autophagy modulation in human islets under conditions associated to endoplasmic reticulum (ER) stress. Human pancreatic islets were isolated by enzymatic digestion and density gradient purification from pancreatic samples of non-diabetic (ND; n = 17; age 65 ± 21 years; gender: 5 M/12 F; BMI 23.4 ± 3.3 kg/m2) and T2D (n = 9; age 76 ± 6 years; 4 M/5 F; gender: BMI 25.4 ± 3.7 kg/m2) organ donors. Nine ND organ donors were treated for hypertension and 1 for both hypertension and hypercholesterolemia. T2D organ donors were treated with metformin (1), oral hypoglycemic agents (2), diet + oral hypoglycemic agents (3), insulin (3) or insulin plus metformin (3) as for antidiabetic therapy and, of these, 3 were treated also for hypertension and 6 for both hypertension and hypercholesterolemia. Two days after isolation, they were cultured for 1-5 days with 10 ng/ml rapamycin (autophagy inducer), 5 mM 3-methyladenine or 1.0 nM concanamycin-A (autophagy blockers), either in the presence or not of metabolic (0.5 mM palmitate) or chemical (0.1 ng/ml brefeldin A) ER stressors. In ND islets palmitate exposure induced a 4 to 5-fold increase of beta cell apoptosis, which was significantly prevented by rapamycin and exacerbated by 3-MA. Similar results were observed with brefeldin treatment. Glucose-stimulated insulin secretion from ND islets was reduced by palmitate (-40 to 50%) and brefeldin (-60 to 70%), and rapamycin counteracted palmitate, but not brefeldin, cytotoxic actions. Both palmitate and brefeldin induced PERK, CHOP and BiP gene expression, which was partially, but significantly prevented by rapamycin. With T2D islets, rapamycin alone reduced the amount of p62, an autophagy receptor that accumulates in cells when macroautophagy is inhibited. Compared to untreated T2D cells, rapamycin-exposed diabetic islets showed improved insulin secretion, reduced proportion of beta cells showing signs of apoptosis and better preserved insulin granules, mitochondria and ER ultrastructure; this was associated with significant reduction of PERK, CHOP and BiP gene expression. This study emphasizes the importance of autophagy modulation in human beta cell function and survival, particularly in situations of ER stress. Tuning autophagy could be a tool for beta cell protection.
Collapse
Affiliation(s)
- M. Bugliani
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - S. Mossuto
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - F. Grano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - M. Suleiman
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - L. Marselli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - U. Boggi
- Department of Surgical Pathology, Medicine, Molecular and Critical Area, University of Pisa, Pisa, Italy
| | - P. De Simone
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - D. L. Eizirik
- Medical Faculty, ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - M. Cnop
- Medical Faculty, ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - P. Marchetti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - V. De Tata
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
- *Correspondence: V. De Tata
| |
Collapse
|
20
|
Groebe K, Cen J, Schvartz D, Sargsyan E, Chowdhury A, Roomp K, Schneider R, Alderborn A, Sanchez JC, Bergsten P. Palmitate-Induced Insulin Hypersecretion and Later Secretory Decline Associated with Changes in Protein Expression Patterns in Human Pancreatic Islets. J Proteome Res 2018; 17:3824-3836. [PMID: 30183308 DOI: 10.1021/acs.jproteome.8b00239] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In obese children with high circulating concentrations of free fatty acid palmitate, we have observed that insulin levels at fasting and in response to a glucose challenge were several times higher than in obese children with low concentrations of the fatty acid as well as in lean controls. Declining and even insufficient insulin levels were observed in obese adolescents with high levels of the fatty acid. In isolated human islets exposed to palmitate we have observed insulin hypersecretion after 2 days exposure. In contrast, insulin secretion from the islets was reduced after 7 days culture in the presence of the fatty acid. This study aims at identifying islet-related biological events potentially linked with the observed insulin hypersecretion and later secretory decline in these obese children and adolescents using the islet model. We analyzed protein expression data obtained from human islets exposed to elevated palmitate levels for 2 and 7 days by an improved methodology for statistical analysis of differentially expressed proteins. Protein profiling of islet samples by liquid chromatography-tandem mass spectrometry identified 115 differentially expressed proteins (DEPs). Several DEPs including sorcin were associated with increased glucose-stimulated insulin secretion in islets after 2 days of exposure to palmitate. Similarly, several metabolic pathways including altered protein degradation, increased autophagy, altered redox condition, and hampered insulin processing were coupled to the functional impairment of islets after 7 days of culture in the presence of palmitate. Such biological events, once validated in the islets, may give rise to novel treatment strategies aiming at normalizing insulin levels in obese children with high palmitate levels, which may reduce or even prevent obesity-related type 2 diabetes mellitus.
Collapse
Affiliation(s)
| | - Jing Cen
- Department of Medical Cell Biology , Uppsala University , 75236 Uppsala , Sweden
| | - Domitille Schvartz
- Human Protein Sciences Department, Centre Medical Universitaire , University of Geneva , CH-1211 Geneva , Switzerland
| | - Ernest Sargsyan
- Department of Medical Cell Biology , Uppsala University , 75236 Uppsala , Sweden
| | - Azazul Chowdhury
- Department of Medical Cell Biology , Uppsala University , 75236 Uppsala , Sweden
| | - Kirsten Roomp
- Luxembourg Centre for Systems Biomedicine , University of Luxembourg , 4365 Esch-sur-Alzette , Luxembourg
| | - Reinhard Schneider
- Luxembourg Centre for Systems Biomedicine , University of Luxembourg , 4365 Esch-sur-Alzette , Luxembourg
| | - Anders Alderborn
- Department of Medical Cell Biology , Uppsala University , 75236 Uppsala , Sweden
| | - Jean-Charles Sanchez
- Human Protein Sciences Department, Centre Medical Universitaire , University of Geneva , CH-1211 Geneva , Switzerland
| | - Peter Bergsten
- Department of Medical Cell Biology , Uppsala University , 75236 Uppsala , Sweden
| |
Collapse
|
21
|
Sargsyan E, Cen J, Roomp K, Schneider R, Bergsten P. Identification of early biological changes in palmitate-treated isolated human islets. BMC Genomics 2018; 19:629. [PMID: 30134843 PMCID: PMC6106933 DOI: 10.1186/s12864-018-5008-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 08/14/2018] [Indexed: 12/13/2022] Open
Abstract
Background Long-term exposure to elevated levels of free fatty acids (FFAs) is deleterious for beta-cell function and may contribute to development of type 2 diabetes mellitus (T2DM). Whereas mechanisms of impaired glucose-stimulated insulin secretion (GSIS) in FFA-treated beta-cells have been intensively studied, biological events preceding the secretory failure, when GSIS is accentuated, are poorly investigated. To identify these early events, we performed genome-wide analysis of gene expression in isolated human islets exposed to fatty acid palmitate for different time periods. Results Palmitate-treated human islets showed decline in beta-cell function starting from day two. Affymetrix Human Transcriptome Array 2.0 identified 903 differentially expressed genes (DEGs). Mapping of the genes onto pathways using KEGG pathway enrichment analysis predicted four islet biology-related pathways enriched prior but not after the decline of islet function and three pathways enriched both prior and after the decline of islet function. DEGs from these pathways were analyzed at the transcript level. The results propose that in palmitate-treated human islets, at early time points, protective events, including up-regulation of metallothioneins, tRNA synthetases and fatty acid-metabolising proteins, dominate over deleterious events, including inhibition of fatty acid detoxification enzymes, which contributes to the enhanced GSIS. After prolonged exposure of islets to palmitate, the protective events are outweighed by the deleterious events, which leads to impaired GSIS. Conclusions The study identifies temporal order between different cellular events, which either promote or protect from beta-cell failure. The sequence of these events should be considered when developing strategies for prevention and treatment of the disease. Electronic supplementary material The online version of this article (10.1186/s12864-018-5008-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ernest Sargsyan
- Department of Medical Cell Biology, Uppsala University, Box 571, 75123, Uppsala, Sweden. .,Molecular Neuroscience Group, Institute of Molecular Biology, National Academy of Sciences, 0014, Yerevan, Armenia.
| | - Jing Cen
- Department of Medical Cell Biology, Uppsala University, Box 571, 75123, Uppsala, Sweden
| | - Kirsten Roomp
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Campus Belval, 7 avenue des Hauts fourneaux, 4362 Esch-Belval, Luxembourg City, Luxembourg
| | - Reinhard Schneider
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Campus Belval, 7 avenue des Hauts fourneaux, 4362 Esch-Belval, Luxembourg City, Luxembourg
| | - Peter Bergsten
- Department of Medical Cell Biology, Uppsala University, Box 571, 75123, Uppsala, Sweden
| |
Collapse
|
22
|
Li Z, Liu H, Niu Z, Zhong W, Xue M, Wang J, Yang F, Zhou Y, Zhou Y, Xu T, Hou J. Temporal Proteomic Analysis of Pancreatic β-Cells in Response to Lipotoxicity and Glucolipotoxicity. Mol Cell Proteomics 2018; 17:2119-2131. [PMID: 30082485 DOI: 10.1074/mcp.ra118.000698] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 08/03/2018] [Indexed: 12/12/2022] Open
Abstract
Chronic hyperlipidemia causes the dysfunction of pancreatic β-cells, such as apoptosis and impaired insulin secretion, which are aggravated in the presence of hyperglycemia. The underlying mechanisms, such as endoplasmic reticulum (ER) stress, oxidative stress and metabolic disorders, have been reported before; however, the time sequence of these molecular events is not fully understood. Here, using isobaric labeling-based mass spectrometry, we investigated the dynamic proteomes of INS-1 cells exposed to high palmitate in the absence and presence of high glucose. Using bioinformatics analysis of differentially expressed proteins, including the time-course expression pattern, protein-protein interaction, gene set enrichment and KEGG pathway analysis, we analyzed the dynamic features of previously reported and newly identified lipotoxicity- and glucolipotoxicity-related molecular events in more detail. Our temporal data highlight cholesterol metabolism occurring at 4 h, earlier than fatty acid metabolism that started at 8 h and likely acting as an early toxic event highly associated with ER stress induced by palmitate. Interestingly, we found that the proliferation of INS-1 cells was significantly increased at 48 h by combined treatment of palmitate and glucose. Moreover, benefit from the time-course quantitative data, we identified and validated two new molecular targets: Setd8 for cell replication and Rhob for apoptosis, demonstrating that our temporal dataset serves as a valuable resource to identify potential candidates for mechanistic studies of lipotoxicity and glucolipotoxicity in pancreatic β-cells.
Collapse
Affiliation(s)
- Zonghong Li
- From the ‡National Laboratory of Biomacramolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.,§Jilin Province Key Laboratory on Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun, 130024, China
| | - Hongyang Liu
- From the ‡National Laboratory of Biomacramolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.,‖Sino-Danish College, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhangjing Niu
- From the ‡National Laboratory of Biomacramolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.,‖Sino-Danish College, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wen Zhong
- ***College of Life Science and Technology, HuaZhong University of Science and Technology, Wuhan 430074, China
| | - Miaomiao Xue
- From the ‡National Laboratory of Biomacramolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.,¶College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jifeng Wang
- ‡‡Laboratory of Protein and Peptide Pharmaceuticals and Laboratory of Proteomics, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Fuquan Yang
- ‡‡Laboratory of Protein and Peptide Pharmaceuticals and Laboratory of Proteomics, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.,¶College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yue Zhou
- §§ThermoFisher Scientific, Building 6, No. 27, Xin Jinqiao Rd, Pudong, Shanghai, 201206, China
| | - Yifa Zhou
- §Jilin Province Key Laboratory on Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun, 130024, China;
| | - Tao Xu
- From the ‡National Laboratory of Biomacramolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; .,¶College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Junjie Hou
- From the ‡National Laboratory of Biomacramolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China;
| |
Collapse
|
23
|
Johnston LW, Harris SB, Retnakaran R, Giacca A, Liu Z, Bazinet RP, Hanley AJ. Association of NEFA composition with insulin sensitivity and beta cell function in the Prospective Metabolism and Islet Cell Evaluation (PROMISE) cohort. Diabetologia 2018; 61:821-830. [PMID: 29275428 DOI: 10.1007/s00125-017-4534-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 11/14/2017] [Indexed: 02/07/2023]
Abstract
AIMS/HYPOTHESIS Our aim was to determine the longitudinal associations of individual NEFA with the pathogenesis of diabetes, specifically with differences in insulin sensitivity and beta cell function over 6 years in a cohort of individuals who are at risk for diabetes. METHODS In the Prospective Metabolism and Islet Cell Evaluation (PROMISE) longitudinal cohort, 477 participants had serum NEFA measured at the baseline visit and completed an OGTT at three time points over 6 years. Outcome variables were calculated using the OGTT values. At each visit, insulin sensitivity was assessed using the HOMA2 of insulin sensitivity (HOMA2-%S) and the Matsuda index, while beta cell function was assessed using the insulinogenic index over HOMA-IR (IGI/IR) and the insulin secretion-sensitivity index-2 (ISSI-2). Generalised estimating equations were used, adjusting for time, waist, sex, ethnicity, baseline age, alanine aminotransferase (ALT) and physical activity. NEFA were analysed as both concentrations (nmol/ml) and proportions (mol%) of the total fraction. RESULTS Participants' (73% female, 70% with European ancestry) insulin sensitivity and beta cell function declined by 14-21% over 6 years of follow-up. In unadjusted models, several NEFA (e.g. 18:1 n-7, 22:4 n-6) were associated with lower insulin sensitivity, however, nearly all of these associations were attenuated in fully adjusted models. In adjusted models, total NEFA, 16:0, 18:1 n-9 and 18:2 n-6 (as concentrations) were associated with 3.7-8.0% lower IGI/IR and ISSI-2, while only 20:5 n-3 (as mol%) was associated with 7.7% higher HOMA2-%S. CONCLUSIONS/INTERPRETATION Total NEFA concentration was a strong predictor of lower beta cell function over 6 years. Our results suggest that the association with beta cell function is due to the absolute size of the serum NEFA fraction, rather than the specific fatty acid composition.
Collapse
Affiliation(s)
- Luke W Johnston
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, FitzGerald Building, 150 College Street, Toronto, ON, M5S 3E2, Canada
| | - Stewart B Harris
- Centre for Studies in Family Medicine, University of Western Ontario, London, ON, Canada
| | - Ravi Retnakaran
- Division of Endocrinology, University of Toronto, Toronto, ON, Canada
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Adria Giacca
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Zhen Liu
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, FitzGerald Building, 150 College Street, Toronto, ON, M5S 3E2, Canada
| | - Richard P Bazinet
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, FitzGerald Building, 150 College Street, Toronto, ON, M5S 3E2, Canada
| | - Anthony J Hanley
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, FitzGerald Building, 150 College Street, Toronto, ON, M5S 3E2, Canada.
- Division of Endocrinology, University of Toronto, Toronto, ON, Canada.
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
24
|
Muilwijk M, Celis-Morales C, Nicolaou M, Snijder MB, Gill JMR, van Valkengoed IGM. Plasma Cholesteryl Ester Fatty Acids do not Mediate the Association of Ethnicity with Type 2 Diabetes: Results From the HELIUS Study. Mol Nutr Food Res 2017; 62. [PMID: 28981995 DOI: 10.1002/mnfr.201700528] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 09/27/2017] [Indexed: 11/12/2022]
Abstract
SCOPE Ethnic minority groups have a higher risk of type 2 diabetes (T2D) than the host population. Our aim is to identify whether plasma cholesteryl ester fatty acids (CEFA) mediate the ethnic differences in type 2 diabetes. METHODS AND RESULTS We included 202 Dutch, 206 South-Asian Surinamese, 205 African Surinamese, 215 Turkish, and 213 Moroccan origin participants of the HELIUS study (Amsterdam, the Netherlands). Logistic regression is used to determine the associations between plasma CEFA and T2D. Mediation analysis is used to identify whether CEFA contributed to the association between ethnicity and T2D. We adjusted for ethnicity, age, sex, smoking, physical activity, and BMI. Associations between plasma CEFA and T2D were similar across all ethnic groups. Although differences in plasma CEFA across ethnic groups were observed, CEFA did not mediate the differences in T2D prevalence between ethnic groups. CONCLUSION Although ethnic differences in plasma CEFA are found and CEFA are associated with T2D, CEFA does not contribute to the difference in T2D prevalence between ethnic groups. If confirmed, this implies that maintenance of the more beneficial CEFA profiles in the non-Dutch ethnic groups may be encouraged to prevent an even higher prevalence of T2D in these groups.
Collapse
Affiliation(s)
- Mirthe Muilwijk
- Department of Public Health, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Carlos Celis-Morales
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Mary Nicolaou
- Department of Public Health, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Marieke B Snijder
- Department of Public Health, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Jason M R Gill
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Irene G M van Valkengoed
- Department of Public Health, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
25
|
Yousefpour A, Modarress H, Goharpey F, Amjad-Iranagh S. Combination of anti-hypertensive drugs: a molecular dynamics simulation study. J Mol Model 2017; 23:158. [DOI: 10.1007/s00894-017-3333-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 03/27/2017] [Indexed: 01/03/2023]
|
26
|
Wang S, Xu L, Lu YT, Liu YF, Han B, Liu T, Tang J, Li J, Wu J, Li JY, Yu LF, Yang F. Discovery of benzofuran-3(2H)-one derivatives as novel DRAK2 inhibitors that protect islet β-cells from apoptosis. Eur J Med Chem 2017; 130:195-208. [PMID: 28249207 DOI: 10.1016/j.ejmech.2017.02.048] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 02/17/2017] [Accepted: 02/18/2017] [Indexed: 12/11/2022]
Abstract
Death-associated protein kinase-related apoptosis-inducing kinase-2 (DRAK2) is a serine/threonine kinase that plays a key role in a wide variety of cell death signaling pathways. Inhibition of DRAK2 was found to efficiently protect islet β-cells from apoptosis and hence DRAK2 inhibitors represent a promising therapeutic strategy for the treatment of diabetes. Only very few chemical entities targeting DRAK2 are currently known. We carried out a high throughput screening and identified compound 4 as a moderate DRAK2 inhibitor with an IC50 value of 3.15 μM. Subsequent SAR studies of hit compound 4 led to the development of novel benzofuran-3(2H)-one series of DRAK2 inhibitors with improved potency and favorable selectivity profiles against 26 selected kinases. Importantly, most potent compounds 40 (IC50 = 0.33 μM) and 41 (IC50 = 0.25 μM) were found to protect islet β-cells from apoptosis in dose-dependent manners. These data support the notion that small molecule inhibitors of DRAK2 represents a promising strategy for the treatment of diabetes.
Collapse
Affiliation(s)
- Sheng Wang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Lei Xu
- Chinese National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 189 Guoshoujing Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
| | - Yu-Ting Lu
- Chinese National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 189 Guoshoujing Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
| | - Yu-Fei Liu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Bing Han
- Laboratory of Immunology and Cardiovascular Research, Centre Hospitalier de l'Université de Montréal, 900 rue St-Denis, Montréal, Québec, Canada
| | - Ting Liu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Jie Tang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China; Shanghai Key Laboratory of Green Chemistry and Chemical Process, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Jia Li
- Chinese National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 189 Guoshoujing Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
| | - Jiangping Wu
- Laboratory of Immunology and Cardiovascular Research, Centre Hospitalier de l'Université de Montréal, 900 rue St-Denis, Montréal, Québec, Canada.
| | - Jing-Ya Li
- Chinese National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 189 Guoshoujing Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China.
| | - Li-Fang Yu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China.
| | - Fan Yang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China.
| |
Collapse
|
27
|
Mukherjee S, Chattopadhyay M, Bhattacharya S, Dasgupta S, Hussain S, Bharadwaj SK, Talukdar D, Usmani A, Pradhan BS, Majumdar SS, Chattopadhyay P, Mukhopadhyay S, Maity TK, Chaudhuri MK, Bhattacharya S. A Small Insulinomimetic Molecule Also Improves Insulin Sensitivity in Diabetic Mice. PLoS One 2017; 12:e0169809. [PMID: 28072841 PMCID: PMC5224995 DOI: 10.1371/journal.pone.0169809] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 12/21/2016] [Indexed: 12/18/2022] Open
Abstract
Dramatic increase of diabetes over the globe is in tandem with the increase in insulin requirement. This is because destruction and dysfunction of pancreatic β-cells are of common occurrence in both Type1 diabetes and Type2 diabetes, and insulin injection becomes a compulsion. Because of several problems associated with insulin injection, orally active insulin mimetic compounds would be ideal substitute. Here we report a small molecule, a peroxyvanadate compound i.e. DmpzH[VO(O2)2(dmpz)], henceforth referred as dmp, which specifically binds to insulin receptor with considerable affinity (KD-1.17μM) thus activating insulin receptor tyrosine kinase and its downstream signaling molecules resulting increased uptake of [14C] 2 Deoxy-glucose. Oral administration of dmp to streptozotocin treated BALB/c mice lowers blood glucose level and markedly stimulates glucose and fatty acid uptake by skeletal muscle and adipose tissue respectively. In db/db mice, it greatly improves insulin sensitivity through excess expression of PPARγ and its target genes i.e. adiponectin, CD36 and aP2. Study on the underlying mechanism demonstrated that excess expression of Wnt3a decreased PPARγ whereas dmp suppression of Wnt3a gene increased PPARγ expression which subsequently augmented adiponectin. Increased production of adiponectin in db/db mice due to dmp effected lowering of circulatory TG and FFA levels, activates AMPK in skeletal muscle and this stimulates mitochondrial biogenesis and bioenergetics. Decrease of lipid load along with increased mitochondrial activity greatly improves energy homeostasis which has been found to be correlated with the increased insulin sensitivity. The results obtained with dmp, therefore, strongly indicate that dmp could be a potential candidate for insulin replacement therapy.
Collapse
Affiliation(s)
- Sandip Mukherjee
- Cellular and Molecular Endocrinology Laboratory, Centre for Advanced Studies in Zoology, School of Life Science, Visva-Bharati (A Central University), Santiniketan, West Bengal, India
| | - Mrittika Chattopadhyay
- Cellular and Molecular Endocrinology Laboratory, Centre for Advanced Studies in Zoology, School of Life Science, Visva-Bharati (A Central University), Santiniketan, West Bengal, India
| | | | - Suman Dasgupta
- Department of Molecular Biology and Biotechnology, Tezpur University, Assam, India
| | - Sahid Hussain
- Department of Chemical Sciences, Tezpur University, Assam, India
| | | | | | - Abul Usmani
- Division of Cellular Endocrinology, National Institute of Immunology, New Delhi, India
| | - Bhola S Pradhan
- Division of Cellular Endocrinology, National Institute of Immunology, New Delhi, India
| | - Subeer S Majumdar
- Division of Cellular Endocrinology, National Institute of Immunology, New Delhi, India
| | | | - Satinath Mukhopadhyay
- Department of Endocrinology & Metabolism, Institute of Post-Graduate Medical Education & Research-Seth Sukhlal Karnani Memorial (IPGME&R−SSKM) Hospital, Kolkata, West Bengal, India
| | | | - Mihir K. Chaudhuri
- Department of Chemical Sciences, Tezpur University, Assam, India
- * E-mail: (SB); (MKC)
| | - Samir Bhattacharya
- Cellular and Molecular Endocrinology Laboratory, Centre for Advanced Studies in Zoology, School of Life Science, Visva-Bharati (A Central University), Santiniketan, West Bengal, India
- * E-mail: (SB); (MKC)
| |
Collapse
|
28
|
Initial hyperinsulinemia and subsequent β-cell dysfunction is associated with elevated palmitate levels. Pediatr Res 2016; 80:267-74. [PMID: 27064244 DOI: 10.1038/pr.2016.80] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 03/03/2016] [Indexed: 12/16/2022]
Abstract
BACKGROUND The prevalence of obesity-related diabetes in childhood is increasing and circulating levels of nonesterified fatty acids may constitute a link. Here, the association between palmitate and insulin secretion was investigated in vivo and in vitro. METHODS Obese and lean children and adolescents (n = 80) were included. Palmitate was measured at fasting; insulin and glucose during an oral glucose tolerance test (OGTT). Human islets were cultured for 0 to 7 d in presence of 0.5 mmol/l palmitate. Glucose-stimulated insulin secretion (GSIS), insulin content and apoptosis were measured. RESULTS Obese subjects had fasting palmitate levels between 0.10 and 0.33 mmol/l, with higher average levels compared to lean subjects. While obese children with elevated palmitate (>0.20 mmol/l) had accentuated insulin levels during OGTT, obese adolescents with high palmitate had delayed first-phase insulin response. In human islets exposed to palmitate for 2 d GSIS was twofold enhanced, but after 7 d attenuated. Intracellular insulin content decreased time-dependently in islets cultured in the presence of palmitate and cleaved caspase 3 increased. CONCLUSION The rapid accentuated and delayed insulin secretory responses observed in obese children and adolescents, respectively, with high palmitate levels may reflect changes in islet secretory activity and integrity induced by extended exposure to the fatty acid.
Collapse
|
29
|
Johnston LW, Harris SB, Retnakaran R, Zinman B, Giacca A, Liu Z, Bazinet RP, Hanley AJ. Longitudinal Associations of Phospholipid and Cholesteryl Ester Fatty Acids With Disorders Underlying Diabetes. J Clin Endocrinol Metab 2016; 101:2536-44. [PMID: 27144932 DOI: 10.1210/jc.2015-4267] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Specific serum fatty acid (FA) profiles predict the development of incident type 2 diabetes; however, limited longitudinal data exist exploring their role in the progression of insulin sensitivity (IS) and β-cell function. OBJECTIVE To examine the longitudinal associations of the FA composition of serum phospholipid (PL) and cholesteryl ester (CE) fractions with IS and β-cell function over 6 years. DESIGN The Prospective Metabolism and Islet Cell Evaluation (PROMISE) cohort is a longitudinal observational study, with clinic visits occurring every 3 years. Three visits have been completed, totaling 6 years of follow-up. SETTING Individuals (n = 477) at risk for diabetes recruited from the general population in London and Toronto, Canada. MAIN OUTCOME MEASURES Values from an oral glucose tolerance test were used to compute 1/HOMA-IR and the Matsuda index for IS, the insulinogenic index over HOMA-IR, and the insulin secretion-sensitivity index-2 for β-cell function. Thin-layer chromatograph and gas chromatograph quantified FA. Generalized estimating equations were used for the analysis. RESULTS IS and β-cell function declined by 8.3-19.4% over 6 years. In fully adjusted generalized estimating equation models, PL cis-vaccenate (18:1n-7) was positively associated with all outcomes, whereas γ-linolenate (GLA; 18:3n-6) and stearate (18:0) were negatively associated with IS. Tests for time interactions revealed that PL eicosadienoate (20:2n-6) and palmitate (16:0) and CE dihomo-γ-linolenate (20:3n-6), GLA, and palmitate had stronger associations with the outcomes after longer follow-up. CONCLUSIONS In a Canadian population at risk for diabetes, we found that higher PL stearate and GLA and lower cis-vaccenic acid predicted consistently lower IS and β-cell function over 6 years.
Collapse
Affiliation(s)
- Luke W Johnston
- Department of Nutritional Sciences (L.W.J., Z.L., R.P.B., A.J.H.), University of Toronto, Toronto, ON M5S 3E2, Canada; Centre for Studies in Family Medicine (S.B.H.), University of Western Ontario, London, ON N6G 2M1, Canada; Division of Endocrinology (R.R., B.Z.), University of Toronto, Toronto, ON M5S 1A8, Canada; Lunenfeld Tanenbaum Research Institute (R.R., B.Z.), Mt Sinai Hospital, Toronto, ON M5T 3L9, Canada; Department of Physiology (A.G.), University of Toronto, Toronto, ON M5S 1A8, Canada; and Dalla Lana School of Public Health (A.J.H.), University of Toronto, Toronto, ON M5T 3M7, Canada
| | - Stewart B Harris
- Department of Nutritional Sciences (L.W.J., Z.L., R.P.B., A.J.H.), University of Toronto, Toronto, ON M5S 3E2, Canada; Centre for Studies in Family Medicine (S.B.H.), University of Western Ontario, London, ON N6G 2M1, Canada; Division of Endocrinology (R.R., B.Z.), University of Toronto, Toronto, ON M5S 1A8, Canada; Lunenfeld Tanenbaum Research Institute (R.R., B.Z.), Mt Sinai Hospital, Toronto, ON M5T 3L9, Canada; Department of Physiology (A.G.), University of Toronto, Toronto, ON M5S 1A8, Canada; and Dalla Lana School of Public Health (A.J.H.), University of Toronto, Toronto, ON M5T 3M7, Canada
| | - Ravi Retnakaran
- Department of Nutritional Sciences (L.W.J., Z.L., R.P.B., A.J.H.), University of Toronto, Toronto, ON M5S 3E2, Canada; Centre for Studies in Family Medicine (S.B.H.), University of Western Ontario, London, ON N6G 2M1, Canada; Division of Endocrinology (R.R., B.Z.), University of Toronto, Toronto, ON M5S 1A8, Canada; Lunenfeld Tanenbaum Research Institute (R.R., B.Z.), Mt Sinai Hospital, Toronto, ON M5T 3L9, Canada; Department of Physiology (A.G.), University of Toronto, Toronto, ON M5S 1A8, Canada; and Dalla Lana School of Public Health (A.J.H.), University of Toronto, Toronto, ON M5T 3M7, Canada
| | - Bernard Zinman
- Department of Nutritional Sciences (L.W.J., Z.L., R.P.B., A.J.H.), University of Toronto, Toronto, ON M5S 3E2, Canada; Centre for Studies in Family Medicine (S.B.H.), University of Western Ontario, London, ON N6G 2M1, Canada; Division of Endocrinology (R.R., B.Z.), University of Toronto, Toronto, ON M5S 1A8, Canada; Lunenfeld Tanenbaum Research Institute (R.R., B.Z.), Mt Sinai Hospital, Toronto, ON M5T 3L9, Canada; Department of Physiology (A.G.), University of Toronto, Toronto, ON M5S 1A8, Canada; and Dalla Lana School of Public Health (A.J.H.), University of Toronto, Toronto, ON M5T 3M7, Canada
| | - Adria Giacca
- Department of Nutritional Sciences (L.W.J., Z.L., R.P.B., A.J.H.), University of Toronto, Toronto, ON M5S 3E2, Canada; Centre for Studies in Family Medicine (S.B.H.), University of Western Ontario, London, ON N6G 2M1, Canada; Division of Endocrinology (R.R., B.Z.), University of Toronto, Toronto, ON M5S 1A8, Canada; Lunenfeld Tanenbaum Research Institute (R.R., B.Z.), Mt Sinai Hospital, Toronto, ON M5T 3L9, Canada; Department of Physiology (A.G.), University of Toronto, Toronto, ON M5S 1A8, Canada; and Dalla Lana School of Public Health (A.J.H.), University of Toronto, Toronto, ON M5T 3M7, Canada
| | - Zhen Liu
- Department of Nutritional Sciences (L.W.J., Z.L., R.P.B., A.J.H.), University of Toronto, Toronto, ON M5S 3E2, Canada; Centre for Studies in Family Medicine (S.B.H.), University of Western Ontario, London, ON N6G 2M1, Canada; Division of Endocrinology (R.R., B.Z.), University of Toronto, Toronto, ON M5S 1A8, Canada; Lunenfeld Tanenbaum Research Institute (R.R., B.Z.), Mt Sinai Hospital, Toronto, ON M5T 3L9, Canada; Department of Physiology (A.G.), University of Toronto, Toronto, ON M5S 1A8, Canada; and Dalla Lana School of Public Health (A.J.H.), University of Toronto, Toronto, ON M5T 3M7, Canada
| | - Richard P Bazinet
- Department of Nutritional Sciences (L.W.J., Z.L., R.P.B., A.J.H.), University of Toronto, Toronto, ON M5S 3E2, Canada; Centre for Studies in Family Medicine (S.B.H.), University of Western Ontario, London, ON N6G 2M1, Canada; Division of Endocrinology (R.R., B.Z.), University of Toronto, Toronto, ON M5S 1A8, Canada; Lunenfeld Tanenbaum Research Institute (R.R., B.Z.), Mt Sinai Hospital, Toronto, ON M5T 3L9, Canada; Department of Physiology (A.G.), University of Toronto, Toronto, ON M5S 1A8, Canada; and Dalla Lana School of Public Health (A.J.H.), University of Toronto, Toronto, ON M5T 3M7, Canada
| | - Anthony J Hanley
- Department of Nutritional Sciences (L.W.J., Z.L., R.P.B., A.J.H.), University of Toronto, Toronto, ON M5S 3E2, Canada; Centre for Studies in Family Medicine (S.B.H.), University of Western Ontario, London, ON N6G 2M1, Canada; Division of Endocrinology (R.R., B.Z.), University of Toronto, Toronto, ON M5S 1A8, Canada; Lunenfeld Tanenbaum Research Institute (R.R., B.Z.), Mt Sinai Hospital, Toronto, ON M5T 3L9, Canada; Department of Physiology (A.G.), University of Toronto, Toronto, ON M5S 1A8, Canada; and Dalla Lana School of Public Health (A.J.H.), University of Toronto, Toronto, ON M5T 3M7, Canada
| |
Collapse
|
30
|
El Ouaamari A, Zhou JY, Liew CW, Shirakawa J, Dirice E, Gedeon N, Kahraman S, De Jesus DF, Bhatt S, Kim JS, Clauss TR, Camp DG, Smith RD, Qian WJ, Kulkarni RN. Compensatory Islet Response to Insulin Resistance Revealed by Quantitative Proteomics. J Proteome Res 2015; 14:3111-3122. [PMID: 26151086 DOI: 10.1021/acs.jproteome.5b00587] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Compensatory islet response is a distinct feature of the prediabetic insulin-resistant state in humans and rodents. To identify alterations in the islet proteome that characterize the adaptive response, we analyzed islets from 5 month old male control, high-fat diet fed (HFD), or obese ob/ob mice by LC-MS/MS and quantified ~1100 islet proteins (at least two peptides) with a false discovery rate < 1%. Significant alterations in abundance were observed for ~350 proteins among groups. The majority of alterations were common to both models, and the changes of a subset of ~40 proteins and 12 proteins were verified by targeted quantification using selected reaction monitoring and western blots, respectively. The insulin-resistant islets in both groups exhibited reduced expression of proteins controlling energy metabolism, oxidative phosphorylation, hormone processing, and secretory pathways. Conversely, an increased expression of molecules involved in protein synthesis and folding suggested effects in endoplasmic reticulum stress response, cell survival, and proliferation in both insulin-resistant models. In summary, we report a unique comparison of the islet proteome that is focused on the compensatory response in two insulin-resistant rodent models that are not overtly diabetic. These data provide a valuable resource of candidate proteins to the scientific community to undertake further studies aimed at enhancing β-cell mass in patients with diabetes. The data are available via the MassIVE repository, under accession no. MSV000079093.
Collapse
Affiliation(s)
- Abdelfattah El Ouaamari
- Islet Cell & Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02215
| | - Jian-Ying Zhou
- Biological Sciences Division and Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99352
| | - Chong Wee Liew
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Jun Shirakawa
- Islet Cell & Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02215
| | - Ercument Dirice
- Islet Cell & Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02215
| | - Nicholas Gedeon
- Islet Cell & Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02215
| | - Sevim Kahraman
- Islet Cell & Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02215
| | - Dario F De Jesus
- Islet Cell & Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02215
| | - Shweta Bhatt
- Islet Cell & Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02215
| | - Jong-Seo Kim
- Biological Sciences Division and Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99352
| | - Therese Rw Clauss
- Biological Sciences Division and Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99352
| | - David G Camp
- Biological Sciences Division and Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99352
| | - Richard D Smith
- Biological Sciences Division and Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99352
| | - Wei-Jun Qian
- Biological Sciences Division and Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99352
| | - Rohit N Kulkarni
- Islet Cell & Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02215
| |
Collapse
|
31
|
Farsi PF, Djazayery A, Eshraghian MR, Koohdani F, Saboor-Yaraghi AA, Derakhshanian H, Zarei M, Javanbakht MH, Djalali M. Effects of supplementation with omega-3 on insulin sensitivity and non-esterified free fatty acid (NEFA) in type 2 diabetic patients. ACTA ACUST UNITED AC 2015; 58:335-40. [PMID: 24936727 DOI: 10.1590/0004-2730000002861] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 10/24/2013] [Indexed: 11/22/2022]
Abstract
OBJECTIVE The aim of this study was to determine the role of omega-3 supplementation on NEFA concentration, insulin sensitivity and resistance, and glucose and lipid metabolism in type 2 diabetic patients. SUBJECTS AND METHODS Forty-four type 2 diabetic patients were randomly recruited into two groups. Group A received 4 g/day omega-3 soft gels, and group B received a placebo for 10 wks. Blood samples were collected after 12-h fast. Physical activity records, three-day food records, and anthropometric measurements were obtained from all participants at the beginning and end of the study. RESULTS Omega-3 supplementation caused a significant reduction in NEFA in the intervention group compared with the placebo group (P = 0.009). Additionally, the administration of omega-3 resulted in significantly greater changes (Diff) for the intervention group in various parameters, such as insulin and Quicki indices compared with the placebo group (P < 0.05). CONCLUSIONS Omega-3 fatty acid supplementation in type 2 diabetic patients improved insulin sensitivity, probably due to the decrease in NEFA concentrations.
Collapse
Affiliation(s)
- Payam Farahbakhsh Farsi
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Abolghassem Djazayery
- Department of Nutrition and Biochemistry, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Eshraghian
- Department of Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Fariba Koohdani
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Akbar Saboor-Yaraghi
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Hoda Derakhshanian
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahnaz Zarei
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hassan Javanbakht
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmoud Djalali
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
32
|
Japtok L, Schmitz EI, Fayyaz S, Krämer S, Hsu LJ, Kleuser B. Sphingosine 1-phosphate counteracts insulin signaling in pancreatic β-cells via the sphingosine 1-phosphate receptor subtype 2. FASEB J 2015; 29:3357-69. [PMID: 25911610 DOI: 10.1096/fj.14-263194] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 04/16/2015] [Indexed: 01/04/2023]
Abstract
Glucolipotoxic stress has been identified as a key player in the progression of pancreatic β-cell dysfunction contributing to insulin resistance and the development of type 2 diabetes mellitus (T2D). It has been suggested that bioactive lipid intermediates, formed under lipotoxic conditions, are involved in these processes. Here, we show that sphingosine 1-phosphate (S1P) levels are not only increased in palmitate-stimulated pancreatic β-cells but also regulate β-cell homeostasis in a divergent manner. Although S1P possesses a prosurvival effect in β-cells, an enhanced level of the sphingolipid antagonizes insulin-mediated cell growth and survival via the sphingosine 1-phosphate receptor subtype 2 (S1P2) followed by an inhibition of Akt-signaling. In an attempt to investigate the role of the S1P/S1P2 axis in vivo, the New Zealand obese (NZO) diabetic mouse model, characterized by β-cell loss under high-fat diet (HFD) conditions, was used. The occurrence of T2D was accompanied by an increase of plasma S1P levels. To examine whether S1P contributes to the morphologic changes of islets via S1P2, the receptor antagonist JTE-013 was administered. Most interestingly, JTE-013 rescued β-cell damage clearly indicating an important role of the S1P2 in β-cell homeostasis. Therefore, the present study provides a new therapeutic strategy to diminish β-cell dysfunction and the development of T2D.
Collapse
Affiliation(s)
- Lukasz Japtok
- *Faculty of Mathematics and Natural Science, Institute of Nutritional Science, Department of Toxicology, University of Potsdam, Potsdam, Germany; German Institute of Human Nutrition, Max Rubner Laboratory, Nuthetal, Germany; and Lpath Incorporated, San Diego, California, USA
| | - Elisabeth I Schmitz
- *Faculty of Mathematics and Natural Science, Institute of Nutritional Science, Department of Toxicology, University of Potsdam, Potsdam, Germany; German Institute of Human Nutrition, Max Rubner Laboratory, Nuthetal, Germany; and Lpath Incorporated, San Diego, California, USA
| | - Susann Fayyaz
- *Faculty of Mathematics and Natural Science, Institute of Nutritional Science, Department of Toxicology, University of Potsdam, Potsdam, Germany; German Institute of Human Nutrition, Max Rubner Laboratory, Nuthetal, Germany; and Lpath Incorporated, San Diego, California, USA
| | - Stephanie Krämer
- *Faculty of Mathematics and Natural Science, Institute of Nutritional Science, Department of Toxicology, University of Potsdam, Potsdam, Germany; German Institute of Human Nutrition, Max Rubner Laboratory, Nuthetal, Germany; and Lpath Incorporated, San Diego, California, USA
| | - Leigh J Hsu
- *Faculty of Mathematics and Natural Science, Institute of Nutritional Science, Department of Toxicology, University of Potsdam, Potsdam, Germany; German Institute of Human Nutrition, Max Rubner Laboratory, Nuthetal, Germany; and Lpath Incorporated, San Diego, California, USA
| | - Burkhard Kleuser
- *Faculty of Mathematics and Natural Science, Institute of Nutritional Science, Department of Toxicology, University of Potsdam, Potsdam, Germany; German Institute of Human Nutrition, Max Rubner Laboratory, Nuthetal, Germany; and Lpath Incorporated, San Diego, California, USA
| |
Collapse
|
33
|
Rondas D, Crèvecoeur I, D'Hertog W, Ferreira GB, Staes A, Garg AD, Eizirik DL, Agostinis P, Gevaert K, Overbergh L, Mathieu C. Citrullinated glucose-regulated protein 78 is an autoantigen in type 1 diabetes. Diabetes 2015; 64:573-86. [PMID: 25204978 DOI: 10.2337/db14-0621] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Posttranslational modifications of self-proteins play a substantial role in the initiation or propagation of the autoimmune attack in several autoimmune diseases, but their contribution to type 1 diabetes is only recently emerging. In the current study, we demonstrate that inflammatory stress, induced by the cytokines interleukin-1β and interferon-γ, leads to citrullination of GRP78 in β-cells. This is coupled with translocation of this endoplasmic reticulum chaperone to the β-cell plasma membrane and subsequent secretion. Importantly, expression and activity of peptidylarginine deiminase 2, one of the five enzymes responsible for citrullination and a candidate gene for type 1 diabetes in mice, is increased in islets from diabetes-prone nonobese diabetic (NOD) mice. Finally, (pre)diabetic NOD mice have autoantibodies and effector T cells that react against citrullinated GRP78, indicating that inflammation-induced citrullination of GRP78 in β-cells generates a novel autoantigen in type 1 diabetes, opening new avenues for biomarker development and therapeutic intervention.
Collapse
Affiliation(s)
- Dieter Rondas
- Laboratory for Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Inne Crèvecoeur
- Laboratory for Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Wannes D'Hertog
- Laboratory for Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | | | - An Staes
- Department of Medical Protein Research, VIB, Ghent, Belgium Department of Biochemistry, Ghent University, Ghent, Belgium
| | - Abhishek D Garg
- Laboratory for Cell Death Research and Therapy, KU Leuven, Leuven, Belgium
| | - Decio L Eizirik
- Laboratory of Experimental Medicine and Université Libre de Bruxelles Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Patrizia Agostinis
- Laboratory for Cell Death Research and Therapy, KU Leuven, Leuven, Belgium
| | - Kris Gevaert
- Department of Medical Protein Research, VIB, Ghent, Belgium Department of Biochemistry, Ghent University, Ghent, Belgium
| | - Lut Overbergh
- Laboratory for Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Chantal Mathieu
- Laboratory for Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| |
Collapse
|
34
|
Ciregia F, Giusti L, Ronci M, Bugliani M, Piga I, Pieroni L, Rossi C, Marchetti P, Urbani A, Lucacchini A. Glucagon-like peptide 1 protects INS-1E mitochondria against palmitate-mediated beta-cell dysfunction: a proteomic study. MOLECULAR BIOSYSTEMS 2015; 11:1696-707. [DOI: 10.1039/c5mb00022j] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Proteomic analysis of the protein expression profiles of enriched mitochondrial preparations of rat INS-1E β cells treated with palmitate in the presence and in the absence of GLP-1.
Collapse
Affiliation(s)
- Federica Ciregia
- Department of Pharmacy
- University of Pisa
- Pisa
- Italy
- Santa Lucia IRCCS Foundation
| | - Laura Giusti
- Department of Pharmacy
- University of Pisa
- Pisa
- Italy
| | - Maurizio Ronci
- Santa Lucia IRCCS Foundation
- Rome
- Italy
- Department of Medical
- Oral and Biotechnological Sciences
| | - Marco Bugliani
- Department of Clinical and Experimental Medicine
- SOD Endocrinology and metabolism of organ and cell transplants-University of Pisa
- Pisa
- Italy
| | | | | | - Claudia Rossi
- Department of Medical
- Oral and Biotechnological Sciences
- University G. d’Annunzio of Chieti-Pescara
- Chieti
- Italy
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine
- SOD Endocrinology and metabolism of organ and cell transplants-University of Pisa
- Pisa
- Italy
| | - Andrea Urbani
- Santa Lucia IRCCS Foundation
- Rome
- Italy
- Department of Experimental Medicine and Surgery
- University of Rome “Tor Vergata”
| | | |
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW β Cells represent one of many cell types in heterogeneous pancreatic islets and play the central role in maintaining glucose homeostasis, such that disrupting β-cell function leads to diabetes. This review summarizes the methods for isolating and characterizing β cells, and describes integrated 'omics' approaches used to define the β cell by its transcriptome and proteome. RECENT FINDINGS RNA sequencing and mass spectrometry-based protein identification have now identified RNA and protein profiles for mouse and human pancreatic islets and β cells, and for β-cell lines. Recent publications have outlined these profiles and, more importantly, have begun to assign the presence or absence of specific genes and regulatory molecules to β-cell function and dysfunction. Overall, researchers have focused on understanding the pathophysiology of diabetes by connecting genome, transcriptome, proteome, and regulatory RNA profiles with findings from genome-wide association studies. SUMMARY Studies employing these relatively new techniques promise to identify specific genes or regulatory RNAs with altered expression as β-cell function begins to deteriorate in the spiral toward the development of diabetes. The ultimate goal is to identify the potential therapeutic targets to prevent β-cell dysfunction and thereby better treat the individual with diabetes. VIDEO ABSTRACT http://links.lww.com/COE/A5.
Collapse
Affiliation(s)
- David M Blodgett
- Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | | | | |
Collapse
|
36
|
Prause M, Christensen DP, Billestrup N, Mandrup-Poulsen T. JNK1 protects against glucolipotoxicity-mediated beta-cell apoptosis. PLoS One 2014; 9:e87067. [PMID: 24475223 PMCID: PMC3901710 DOI: 10.1371/journal.pone.0087067] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 12/23/2013] [Indexed: 12/20/2022] Open
Abstract
Pancreatic β-cell dysfunction is central to type 2 diabetes pathogenesis. Prolonged elevated levels of circulating free-fatty acids and hyperglycemia, also termed glucolipotoxicity, mediate β-cell dysfunction and apoptosis associated with increased c-Jun N-terminal Kinase (JNK) activity. Endoplasmic reticulum (ER) and oxidative stress are elicited by palmitate and high glucose concentrations further potentiating JNK activity. Our aim was to determine the role of the JNK subtypes JNK1, JNK2 and JNK3 in palmitate and high glucose-induced β-cell apoptosis. We established insulin-producing INS1 cell lines stably expressing JNK subtype specific shRNAs to understand the differential roles of the individual JNK isoforms. JNK activity was increased after 3 h of palmitate and high glucose exposure associated with increased expression of ER and mitochondrial stress markers. JNK1 shRNA expressing INS1 cells showed increased apoptosis and cleaved caspase 9 and 3 compared to non-sense shRNA expressing control INS1 cells when exposed to palmitate and high glucose associated with increased CHOP expression, ROS formation and Puma mRNA expression. JNK2 shRNA expressing INS1 cells did not affect palmitate and high glucose induced apoptosis or ER stress markers, but increased Puma mRNA expression compared to non-sense shRNA expressing INS1 cells. Finally, JNK3 shRNA expressing INS1 cells did not induce apoptosis compared to non-sense shRNA expressing INS1 cells when exposed to palmitate and high glucose but showed increased caspase 9 and 3 cleavage associated with increased DP5 and Puma mRNA expression. These data suggest that JNK1 protects against palmitate and high glucose-induced β-cell apoptosis associated with reduced ER and mitochondrial stress.
Collapse
Affiliation(s)
- Michala Prause
- Endocrinology Research Section, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- * E-mail:
| | - Dan Ploug Christensen
- Endocrinology Research Section, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nils Billestrup
- Section of Cellular and Metabolic Research, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Mandrup-Poulsen
- Endocrinology Research Section, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
37
|
Lipidomic profiling before and after Roux-en-Y gastric bypass in obese patients with diabetes. THE PHARMACOGENOMICS JOURNAL 2013; 14:201-7. [PMID: 24365785 DOI: 10.1038/tpj.2013.42] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 11/05/2013] [Accepted: 11/12/2013] [Indexed: 01/11/2023]
Abstract
Bariatric surgery is a well-established approach to improve metabolic disease in morbidly obese patients with high cardiovascular risk. The post-operative normalization of lipid metabolism has a central role in the prevention of future cardiovascular events. The aim of the present study therefore was to characterize changes of plasma lipidomic patterns, consisting of 229 lipid species of 13 lipid classes, 3 months after Roux-en-Y gastric bypass (RYGB) in morbidly obese patients with and without diabetes. RYGB resulted in a 15-32% decrease of body mass index, which was associated with a significant reduction of total cholesterol (TC, -28.3%; P=0.02), LDL-cholesterol (LDL-C, -26.8%; P=0.03) and triglycerides (TGs, -63.0%; P=0.05) measured by routine clinical chemistry. HDL-cholesterol remained unchanged. The effect of RYGB on the plasma lipidomic profile was characterized by significant decreases of 87 lipid species from triacylglycerides (TAGs), cholesterol esters (CholEs), lysophosphatidylcholines (LPCs), phosphatidylcholines (PCs), phosphatidylethanolamine ethers (PEOs), phosphatidylinositols (PIs) and ceramides (Cers). The total of plasma lipid components exhibited a substantial decline of 32.6% and 66 lipid species showed a decrease by over 50%. A direct correlation with HbA1C values could be demonstrated for 24 individual lipid species (10 TAG, three CholE, two LPC, one lysophosphatidylcholine ethers (LPCO) (LPC ether), one PC, two phosphatidylcholine ethers (PCO) and five Cer). Notably, two lipid species (TAG 58:5 and PEO 40:5) were inversely correlated with HbA1C. LPCO, as single whole lipid class, was directly related to HbA1C. These data indicate that RYGB-induced modulation of lipidomic profiles provides important information about post-operative metabolic adaptations and might substantially contribute to improvements of glycemic control. These striking changes in the human plasma lipidome may explain acute, weight independent and long-term effects of RYGB on the cardiovascular system, mental status and immune regulation.
Collapse
|
38
|
Brun T, Scarcia P, Li N, Gaudet P, Duhamel D, Palmieri F, Maechler P. Changes in mitochondrial carriers exhibit stress-specific signatures in INS-1Eβ-cells exposed to glucose versus fatty acids. PLoS One 2013; 8:e82364. [PMID: 24349266 PMCID: PMC3861392 DOI: 10.1371/journal.pone.0082364] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 10/22/2013] [Indexed: 11/19/2022] Open
Abstract
Chronic exposure of β-cells to metabolic stresses impairs their function and potentially induces apoptosis. Mitochondria play a central role in coupling glucose metabolism to insulin secretion. However, little is known on mitochondrial responses to specific stresses; i.e. low versus high glucose, saturated versus unsaturated fatty acids, or oxidative stress. INS-1E cells were exposed for 3 days to 5.6 mM glucose, 25 mM glucose, 0.4 mM palmitate, and 0.4 mM oleate. Culture at standard 11.1 mM glucose served as no-stress control and transient oxidative stress (200 µM H2O2 for 10 min at day 0) served as positive stressful condition. Mito-array analyzed transcripts of 60 mitochondrion-associated genes with special focus on members of the Slc25 family. Transcripts of interest were evaluated at the protein level by immunoblotting. Bioinformatics analyzed the expression profiles to delineate comprehensive networks. Chronic exposure to the different metabolic stresses impaired glucose-stimulated insulin secretion; revealing glucotoxicity and lipo-dysfunction. Both saturated and unsaturated fatty acids increased expression of the carnitine/acylcarnitine carrier CAC, whereas the citrate carrier CIC and energy sensor SIRT1 were specifically upregulated by palmitate and oleate, respectively. High glucose upregulated CIC, the dicarboxylate carrier DIC and glutamate carrier GC1. Conversely, it reduced expression of energy sensors (AMPK, SIRT1, SIRT4), metabolic genes, transcription factor PDX1, and anti-apoptotic Bcl2. This was associated with caspase-3 cleavage and cell death. Expression levels of GC1 and SIRT4 exhibited positive and negative glucose dose-response, respectively. Expression profiles of energy sensors and mitochondrial carriers were selectively modified by the different conditions, exhibiting stress-specific signatures.
Collapse
Affiliation(s)
- Thierry Brun
- Department of Cell Physiology and Metabolism, University of Geneva, Medical Center, Geneva, Switzerland
- * E-mail: (TB); (PM)
| | - Pasquale Scarcia
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Ning Li
- Department of Cell Physiology and Metabolism, University of Geneva, Medical Center, Geneva, Switzerland
| | - Pascale Gaudet
- Swiss Institute of Bioinformatics (SIB) and University of Geneva, Medical Center, Geneva, Switzerland
| | - Dominique Duhamel
- Department of Cell Physiology and Metabolism, University of Geneva, Medical Center, Geneva, Switzerland
| | - Ferdinando Palmieri
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
- Center of Excellence in Comparative Genomics (CEGBA), University of Bari, Bari, Italy
| | - Pierre Maechler
- Department of Cell Physiology and Metabolism, University of Geneva, Medical Center, Geneva, Switzerland
- * E-mail: (TB); (PM)
| |
Collapse
|
39
|
Mazuy C, Ploton M, Eeckhoute J, Berrabah W, Staels B, Lefebvre P, Helleboid-Chapman A. Palmitate increases Nur77 expression by modulating ZBP89 and Sp1 binding to the Nur77 proximal promoter in pancreatic β-cells. FEBS Lett 2013; 587:S0014-5793(13)00781-3. [PMID: 24512852 DOI: 10.1016/j.febslet.2013.10.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 09/18/2013] [Accepted: 10/15/2013] [Indexed: 11/23/2022]
Abstract
Nur77 is a stress sensor in pancreatic β-cells, which negatively regulates glucose-stimulated insulin secretion. We recently showed that a lipotoxic shock caused by exposure of β-cells to the saturated fatty acid palmitate strongly increases Nur77 expression. Here, using dual luciferase reporter assays and Nur77 promoter deletion constructs, we identified a regulatory cassette between -1534 and -1512 bp upstream from the translational start site mediating Nur77 promoter activation in response to palmitate exposure. Chromatin immunoprecipitation, transient transfection and siRNA-mediated knockdown assays revealed that palmitate induced Nur77 promoter activation involves Sp1 recruitment and ZBP89 release from the gene promoter.
Collapse
Affiliation(s)
- Claire Mazuy
- European Genomic Institute for Diabetes (EGID), FR 3508, F-59000 Lille, France; UNIV LILLE 2, F-59000 Lille, France; Inserm UMR 1011, F-59000 Lille, France; IPL, F-59000 Lille, France
| | - Maheul Ploton
- European Genomic Institute for Diabetes (EGID), FR 3508, F-59000 Lille, France; UNIV LILLE 2, F-59000 Lille, France; Inserm UMR 1011, F-59000 Lille, France; IPL, F-59000 Lille, France
| | - Jérôme Eeckhoute
- European Genomic Institute for Diabetes (EGID), FR 3508, F-59000 Lille, France; UNIV LILLE 2, F-59000 Lille, France; Inserm UMR 1011, F-59000 Lille, France; IPL, F-59000 Lille, France
| | - Wahiba Berrabah
- European Genomic Institute for Diabetes (EGID), FR 3508, F-59000 Lille, France; UNIV LILLE 2, F-59000 Lille, France; Inserm UMR 1011, F-59000 Lille, France; IPL, F-59000 Lille, France
| | - Bart Staels
- European Genomic Institute for Diabetes (EGID), FR 3508, F-59000 Lille, France; UNIV LILLE 2, F-59000 Lille, France; Inserm UMR 1011, F-59000 Lille, France; IPL, F-59000 Lille, France
| | - Philippe Lefebvre
- European Genomic Institute for Diabetes (EGID), FR 3508, F-59000 Lille, France; UNIV LILLE 2, F-59000 Lille, France; Inserm UMR 1011, F-59000 Lille, France; IPL, F-59000 Lille, France
| | - Audrey Helleboid-Chapman
- European Genomic Institute for Diabetes (EGID), FR 3508, F-59000 Lille, France; UNIV LILLE 2, F-59000 Lille, France; Inserm UMR 1011, F-59000 Lille, France; IPL, F-59000 Lille, France
| |
Collapse
|