101
|
Moullé VS, Ghislain J, Poitout V. Nutrient regulation of pancreatic β-cell proliferation. Biochimie 2017; 143:10-17. [PMID: 28987628 DOI: 10.1016/j.biochi.2017.09.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 09/28/2017] [Indexed: 12/17/2022]
Abstract
Excess consumption of energy-dense foods combined with a sedentary lifestyle is driving an obesity epidemic. Although obesity is closely associated with insulin resistance, most individuals meet the insulin demand by increasing their functional β-cell mass. Those who eventually develop type 2 diabetes are distinguished by a failure in this compensatory process. Although a causal role of insulin resistance in compensatory β-cell responses has received considerable experimental support, precisely how the β cell senses changes in the metabolic environment is still unknown. As metabolism of glucose, lipids and amino acids is profoundly altered in obesity, it is not surprising that these nutrients are conspicuous among the factors proposed to contribute. In this review we summarise our understanding of the role of nutrients, in particular glucose, fatty acids and amino acids in β-cell compensation with a particular emphasis on their relation to insulin resistance-induced factors and their underlying mechanism of action. Finally, we describe the concept of epigenetic programming and review recent studies illustrating how the status of the β cell epigenome is a product of its nutrient environment, and how metabolic programming of the β cell contributes to diabetes risk.
Collapse
Affiliation(s)
- Valentine S Moullé
- Montreal Diabetes Research Center, University of Montreal, QC, Canada; CRCHUM, University of Montreal, QC, Canada.
| | - Julien Ghislain
- Montreal Diabetes Research Center, University of Montreal, QC, Canada; CRCHUM, University of Montreal, QC, Canada.
| | - Vincent Poitout
- Montreal Diabetes Research Center, University of Montreal, QC, Canada; CRCHUM, University of Montreal, QC, Canada; Department of Medicine, University of Montreal, QC, Canada; Department of Biochemistry and Molecular Medicine, University of Montreal, QC, Canada.
| |
Collapse
|
102
|
Lamontagne J, Al-Mass A, Nolan CJ, Corkey BE, Madiraju SRM, Joly E, Prentki M. Identification of the signals for glucose-induced insulin secretion in INS1 (832/13) β-cells using metformin-induced metabolic deceleration as a model. J Biol Chem 2017; 292:19458-19468. [PMID: 28972173 DOI: 10.1074/jbc.m117.808105] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 09/13/2017] [Indexed: 12/23/2022] Open
Abstract
Metabolic deceleration in pancreatic β-cells is associated with inhibition of glucose-induced insulin secretion (GIIS), but only in the presence of intermediate/submaximal glucose concentrations. Here, we used acute metformin treatment as a tool to induce metabolic deceleration in INS1 (832/13) β-cells, with the goal of identifying key pathways and metabolites involved in GIIS. Metabolites and pathways previously implicated as signals for GIIS were measured in the cells at 2-25 mm glucose, with or without 5 mm metformin. We defined three criteria to identify candidate signals: 1) glucose-responsiveness, 2) sensitivity to metformin-induced inhibition of the glucose effect at intermediate glucose concentrations, and 3) alleviation of metformin inhibition by elevated glucose concentrations. Despite the lack of recovery from metformin-induced impairment of mitochondrial energy metabolism (glucose oxidation, O2 consumption, and ATP production), insulin secretion was almost completely restored at elevated glucose concentrations. Meeting the criteria for candidates involved in promoting GIIS were the following metabolic indicators and metabolites: cytosolic NAD+/NADH ratio (inferred from the dihydroxyacetone phosphate:glycerol-3-phosphate ratio), mitochondrial membrane potential, ADP, Ca2+, 1-monoacylglycerol, diacylglycerol, malonyl-CoA, and HMG-CoA. On the contrary, most of the purine and nicotinamide nucleotides, acetoacetyl-CoA, H2O2, reduced glutathione, and 2-monoacylglycerol were not glucose-responsive. Overall these results underscore the significance of mitochondrial energy metabolism-independent signals in GIIS regulation; in particular, the candidate lipid signaling molecules 1-monoacylglycerol, diacylglycerol, and malonyl-CoA; the predominance of KATP/Ca2+ signaling control by low ADP·Mg2+ rather than by high ATP levels; and a role for a more oxidized state (NAD+/NADH) in the cytosol during GIIS that favors high glycolysis rates.
Collapse
Affiliation(s)
- Julien Lamontagne
- From the Molecular Nutrition Unit and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec H2X 0A9, Canada
| | - Anfal Al-Mass
- From the Molecular Nutrition Unit and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec H2X 0A9, Canada.,the Department of Medicine, McGill University, Montréal, Québec H4A 3J1, Canada
| | - Christopher J Nolan
- the Department of Endocrinology, Canberra Hospital and the Medical School, Australian National University, Canberra ACT 2605, Australia, and
| | - Barbara E Corkey
- the Department of Medicine, Obesity Research Center, Boston University School of Medicine, Boston, Massachusetts 02118
| | - S R Murthy Madiraju
- From the Molecular Nutrition Unit and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec H2X 0A9, Canada
| | - Erik Joly
- From the Molecular Nutrition Unit and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec H2X 0A9, Canada
| | - Marc Prentki
- From the Molecular Nutrition Unit and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec H2X 0A9, Canada, .,the Departments of Nutrition and Biochemistry, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| |
Collapse
|
103
|
Mondal P, Prasad A, Girdhar K. Interventions to improve β-cell mass and function. ANNALES D'ENDOCRINOLOGIE 2017; 78:469-477. [PMID: 28870707 DOI: 10.1016/j.ando.2016.11.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 10/18/2016] [Accepted: 11/09/2016] [Indexed: 01/09/2023]
Abstract
Diabetes mellitus (T2DM) has become an epidemiologically important disease worldwide and is also becoming a great matter of concern due to the effects associated with it like: high morbidity, elevated health care cost and shortened life span. T2DM is a chronic metabolic disease characterized by insulin resistance as well as β-cell dysfunction. It is widely accepted that in the face of insulin resistance, euglycemia can be maintained by increase in pancreatic β-cell mass and insulin secretion. This compensation is largely due to enhanced secretion of insulin by the β-cell mass, which is present initially, and thereby subsequent increases in β-cell mass provide additional insulin secretion. However, the mechanism by which β-cell anatomical plasticity and functional plasticity for insulin secretion is coordinated and executed in different physiological and pathophysiological states is complex and has been poorly understood. As the incidence of T2DM continues to increase at an alarming rate, it is becoming imperative to shift the research focus towards the β-cell physiology where identification of novel pathways that influence the β-cell proliferation and/or contribute to increase insulin secretion has the potential to lead to new therapies for preventing or delaying onset of disease.
Collapse
Affiliation(s)
- Prosenjit Mondal
- School of Basic Sciences, BioX, Indian Institute of Technology, Mandi, HP 175005, India.
| | - Amit Prasad
- School of Basic Sciences, BioX, Indian Institute of Technology, Mandi, HP 175005, India
| | - Khyati Girdhar
- School of Basic Sciences, BioX, Indian Institute of Technology, Mandi, HP 175005, India
| |
Collapse
|
104
|
Brelje TC, Bhagroo NV, Stout LE, Sorenson RL. Prolactin and oleic acid synergistically stimulate β-cell proliferation and growth in rat islets. Islets 2017; 9:e1330234. [PMID: 28686504 PMCID: PMC5510617 DOI: 10.1080/19382014.2017.1330234] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Islet adaptation to pregnancy is largely influenced by prolactin and placental lactogens. In addition serum lipids are significantly increased. Here, we report the novel observation that prolactin and oleic acid synergistically stimulate islet cell proliferation and islet growth. In neonatal rat islets, prolactin increased proliferation 6-fold, oleic acid 3.5-fold, and their combination 15-fold. The expression of insulin in these dividing cells establishes them as β-cells. Similar changes were seen in islet growth. This synergy is restricted to monounsaturated fatty acids and does not occur with other islet growth factors. Oleic acid increases prolactin-induced STAT5 phosphorylation, even though by itself it is unable to induce STAT5 phosphorylation. Their effects on Erk1/2 phosphorylation are additive. Some of the synergy requires the formation of oleoyl CoA and/or its metabolites. Unexpectedly, methyl oleic acid, a non-metabolizable analog of oleic acid, also shows synergy with prolactin. In summary, prolactin and oleic acid synergistically stimulate islet cell proliferation and islet growth in rat islets, oleic acid increases prolactin-induced STAT5 activation, and requires both the metabolism of oleic acid and non-metabolized oleic acid. Since oleic acid is the most abundant monounsaturated fatty acid in serum that is elevated during pregnancy, it may contribute to increased β-cell proliferation seen during pregnancy.
Collapse
Affiliation(s)
- Todd Clark Brelje
- Department of Genetics, Cell Biology, and Development, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Nicholas V. Bhagroo
- Department of Genetics, Cell Biology, and Development, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Laurence E. Stout
- Department of Genetics, Cell Biology, and Development, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Robert L. Sorenson
- Department of Genetics, Cell Biology, and Development, University of Minnesota Medical School, Minneapolis, MN, USA
- CONTACT Robert L. Sorenson Department of Genetics, Cell Biology, and Development, University of Minnesota Medical School, 6–160 Jackson Hall, 321 Church Street SE, Minneapolis, MN 55455, USA
| |
Collapse
|
105
|
Pantophlet AJ, Roelofsen H, de Vries MP, Gerrits WJJ, van den Borne JJGC, Vonk RJ. The use of metabolic profiling to identify insulin resistance in veal calves. PLoS One 2017; 12:e0179612. [PMID: 28617863 PMCID: PMC5472311 DOI: 10.1371/journal.pone.0179612] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 05/31/2017] [Indexed: 11/30/2022] Open
Abstract
Heavy veal calves (4–6 months old) are at risk of developing insulin resistance and disturbed glucose homeostasis. Prolonged insulin resistance could lead to metabolic disorders and impaired growth performance. Recently, we discovered that heavy Holstein-Friesian calves raised on a high-lactose or high-fat diet did not differ in insulin sensitivity, that insulin sensitivity was low and 50% of the calves could be considered insulin resistant. Understanding the patho-physiological mechanisms underlying insulin resistance and discovering biomarkers for early diagnosis would be useful for developing prevention strategies. Therefore, we explored plasma metabolic profiling techniques to build models and discover potential biomarkers and pathways that can distinguish between insulin resistant and moderately insulin sensitive veal calves. The calves (n = 14) were classified as insulin resistant (IR) or moderately insulin sensitive (MIS) based on results from a euglycemic-hyperinsulinemic clamp, using a cut-off value (M/I-value <4.4) to identify insulin resistance. Metabolic profiles of fasting plasma samples were analyzed using reversed phase (RP) and hydrophilic interaction (HILIC) liquid chromatography–mass spectrometry (LC-MS). Orthogonal partial least square discriminant analysis was performed to compare metabolic profiles. Insulin sensitivity was on average 2.3x higher (P <0.001) in MIS than IR group. For both RP-LC-MS and HILIC-LC-MS satisfactory models were build (R2Y >90% and Q2Y >66%), which allowed discrimination between MIS and IR calves. A total of 7 and 20 metabolic features (for RP-LC-MS and HILIC-LC-MS respectively) were most responsible for group separation. Of these, 7 metabolites could putatively be identified that differed (P <0.05) between groups (potential biomarkers). Pathway analysis indicated disturbances in glycerophospholipid and sphingolipid metabolism, the glycine, serine and threonine metabolism, and primary bile acid biosynthesis. These results demonstrate that plasma metabolic profiling can be used to identify insulin resistance in veal calves and can lead to underlying mechanisms.
Collapse
Affiliation(s)
- Andre J Pantophlet
- Department of Pediatrics; Center for Liver, Digestive and Metabolic Diseases, University Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Han Roelofsen
- Medical Biomics, University Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Marcel P de Vries
- Department of Pediatrics; Center for Liver, Digestive and Metabolic Diseases, University Groningen, University Medical Centre Groningen, Groningen, The Netherlands.,Medical Biomics, University Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Walter J J Gerrits
- Animal Nutrition Group, Wageningen University, Wageningen, The Netherlands
| | | | - Roel J Vonk
- Medical Biomics, University Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| |
Collapse
|
106
|
Nurdiana S, Goh YM, Ahmad H, Dom SM, Syimal’ain Azmi N, Noor Mohamad Zin NS, Ebrahimi M. Changes in pancreatic histology, insulin secretion and oxidative status in diabetic rats following treatment with Ficus deltoidea and vitexin. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 17:290. [PMID: 28576138 PMCID: PMC5457635 DOI: 10.1186/s12906-017-1762-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 04/28/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND The potential application of Ficus deltoidea and vitexin for the management of symptomatologies associated with diabetes mellitus (DM) has gained much attention. However, less firm evidence comes from data to augment our understanding of the role of F. deltoidea and vitexin in protecting pancreatic β-cells. The aim of this study was to assess histological and oxidative stress changes in the pancreas of streptozotocin (STZ)-induced diabetic rats following F. deltoidea extract and vitexin treatment. METHODS F. deltoidea and vitexin was administrated orally to six-weeks STZ-induced diabetic rats over 8 weeks period. The glucose and insulin tolerances were assessed by intraperitoneal glucose (2 g/kg) tolerance test (IPGTT) and intraperitoneal insulin (0.65 U/kg) tolerance test (IPITT), respectively. Subsequently, insulin resistance was assessed by homeostasis assessment model of insulin resistance (HOMA-IR), quantitative insulin sensitivity check index (QUICKI) and the insulin/triglyceride-derived McAuley index. The histological changes in the pancreas were then observed by hematoxylin-eosin (H&E) staining. Further, the pattern of fatty acid composition and infrared (IR) spectra of the serum and pancreas were monitored by gas chromatography (GC) method and Fourier Transform Infrared (FT-IR) spectroscopy. RESULTS F. deltoidea and vitexin increased pancreatic antioxidant enzymes and promoted islet regeneration. However, a significant increase in insulin secretion was observed only in rats treated with F. deltoidea. More importantly, reduction of fasting blood glucose is consistent with reduced FT-IR peaks at 1200-1000 cm-1. CONCLUSIONS These results accentuate that F. deltoidea and vitexin could be a potential agent to attenuate pancreatic oxidative damage and advocate their therapeutic potential for treating DM.
Collapse
|
107
|
Abstract
The pancreatic β-cell secretes insulin in response to elevated plasma glucose. This review applies an external bioenergetic critique to the central processes of glucose-stimulated insulin secretion, including glycolytic and mitochondrial metabolism, the cytosolic adenine nucleotide pool, and its interaction with plasma membrane ion channels. The control mechanisms responsible for the unique responsiveness of the cell to glucose availability are discussed from bioenergetic and metabolic control standpoints. The concept of coupling factor facilitation of secretion is critiqued, and an attempt is made to unravel the bioenergetic basis of the oscillatory mechanisms controlling secretion. The need to consider the physiological constraints operating in the intact cell is emphasized throughout. The aim is to provide a coherent pathway through an extensive, complex, and sometimes bewildering literature, particularly for those unfamiliar with the field.
Collapse
Affiliation(s)
- David G Nicholls
- Buck Institute for Research on Aging, Novato, California; and Department of Clinical Sciences, Unit of Molecular Metabolism, Lund University Diabetes Centre, Malmo, Sweden
| |
Collapse
|
108
|
Nie L, Shuai L, Zhu M, Liu P, Xie ZF, Jiang S, Jiang HW, Li J, Zhao Y, Li JY, Tan M. The Landscape of Histone Modifications in a High-Fat Diet-Induced Obese (DIO) Mouse Model. Mol Cell Proteomics 2017; 16:1324-1334. [PMID: 28450421 DOI: 10.1074/mcp.m117.067553] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 04/20/2017] [Indexed: 12/24/2022] Open
Abstract
Type 2 diabetes (T2D) is a major chronic healthcare concern worldwide. Emerging evidence suggests that a histone-modification-mediated epigenetic mechanism underlies T2D. Nevertheless, the dynamics of histone marks in T2D have not yet been carefully analyzed. Using a mass spectrometry-based label-free and chemical stable isotope labeling quantitative proteomic approach, we systematically profiled liver histone post-translational modifications (PTMs) in a prediabetic high-fat diet-induced obese (DIO) mouse model. We identified 170 histone marks, 30 of which were previously unknown. Interestingly, about 30% of the histone marks identified in DIO mouse liver belonged to a set of recently reported lysine acylation modifications, including propionylation, butyrylation, malonylation, and succinylation, suggesting possible roles of these newly identified histone acylations in diabetes and obesity. These histone marks were detected without prior affinity enrichment with an antibody, demonstrating that the histone acylation marks are present at reasonably high stoichiometry. Fifteen histone marks differed in abundance in DIO mouse liver compared with liver from chow-fed mice in label-free quantification, and six histone marks in stable isotope labeling quantification. Analysis of hepatic histone modifications from metformin-treated DIO mice revealed that metformin, a drug widely used for T2D, could reverse DIO-stimulated histone H3K36me2 in prediabetes, suggesting that this mark is likely associated with T2D development. Our study thus offers a comprehensive landscape of histone marks in a prediabetic mouse model, provides a resource for studying epigenetic functions of histone modifications in obesity and T2D, and suggest a new epigenetic mechanism for the physiological function of metformin.
Collapse
Affiliation(s)
- Litong Nie
- From the ‡The Chemical Proteomics Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China, 201203.,§State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China, 201203.,¶University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Lin Shuai
- §State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China, 201203.,¶University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Mingrui Zhu
- From the ‡The Chemical Proteomics Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China, 201203.,§State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China, 201203.,¶University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Ping Liu
- From the ‡The Chemical Proteomics Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China, 201203.,§State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China, 201203
| | - Zhi-Fu Xie
- §State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China, 201203.,¶University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Shangwen Jiang
- From the ‡The Chemical Proteomics Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China, 201203.,§State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China, 201203
| | - Hao-Wen Jiang
- ‖College of Chemistry and Molecular Engineering, East China Normal University, China, 200062
| | - Jia Li
- §State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China, 201203
| | - Yingming Zhao
- From the ‡The Chemical Proteomics Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China, 201203.,§State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China, 201203.,**Ben May Department for Cancer Research, University of Chicago, Chicago, IL
| | - Jing-Ya Li
- §State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China, 201203;
| | - Minjia Tan
- From the ‡The Chemical Proteomics Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China, 201203; .,§State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China, 201203
| |
Collapse
|
109
|
Lutz SZ, Ullrich A, Häring HU, Ullrich S, Gerst F. Sunitinib specifically augments glucose-induced insulin secretion. Cell Signal 2017; 36:91-97. [PMID: 28449948 DOI: 10.1016/j.cellsig.2017.04.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 04/07/2017] [Accepted: 04/23/2017] [Indexed: 01/01/2023]
Abstract
The tyrosine kinase inhibitor sunitinib is used for the treatment of numerous cancers in humans. In diabetic patients, sunitinib lowers blood glucose levels and improves glycaemic control. This study aims to analyse whether sunitinib has specific and direct effects on insulin secreting β-cells. Regulation of insulin secretion, of cellular cAMP levels and activation of signalling pathways were examined upon exposure of rat insulinoma INS-1E cells to sunitinib under specific stimulatory and inhibitory conditions. Secreted insulin and cellular cAMP levels were measured using RIA and ELISA, respectively. Protein phosphorylations were examined on western blots. Sunitinib enhanced glucose-induced insulin secretion (GIIS) concentration-dependently, reaching a maximal stimulation at 2μM. Sunitinib further augmented insulin secretion in the presence of elevated cAMP levels and the FFAR1 agonists. Adrenaline and the PKA inhibitor H89 counteracted the stimulatory effect of sunitinib on secretion. However, sunitinib altered neither the cellular levels of cAMP nor the phosphorylation of PKA. Sunitinib did not reduce IGF-1-induced phosphorylation of AKT/PKB and ERK1/2. In conclusion, these results suggest that sunitinib stimulates GIIS by a direct effect on β-cells, which may contribute to the glucose-lowering action of the tyrosine kinase inhibitor in humans.
Collapse
Affiliation(s)
- Stefan Z Lutz
- German Center for Diabetes Research (DZD e.V.), Germany; Institute for Diabetes Research and Metabolic Diseases IDM of the Helmholtz Center Munich at the Eberhard-Karls-University of Tübingen, Germany; University Hospital Tübingen, Internal Medicine IV, Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Otfried-Müller-Str. 10, 72076 Tübingen, Germany
| | - Axel Ullrich
- Department of Molecular Biology, Max-Planck-Institute of Biochemistry, Martinsried, Germany
| | - Hans-Ulrich Häring
- German Center for Diabetes Research (DZD e.V.), Germany; Institute for Diabetes Research and Metabolic Diseases IDM of the Helmholtz Center Munich at the Eberhard-Karls-University of Tübingen, Germany; University Hospital Tübingen, Internal Medicine IV, Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Otfried-Müller-Str. 10, 72076 Tübingen, Germany
| | - Susanne Ullrich
- German Center for Diabetes Research (DZD e.V.), Germany; Institute for Diabetes Research and Metabolic Diseases IDM of the Helmholtz Center Munich at the Eberhard-Karls-University of Tübingen, Germany; University Hospital Tübingen, Internal Medicine IV, Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Otfried-Müller-Str. 10, 72076 Tübingen, Germany
| | - Felicia Gerst
- German Center for Diabetes Research (DZD e.V.), Germany; Institute for Diabetes Research and Metabolic Diseases IDM of the Helmholtz Center Munich at the Eberhard-Karls-University of Tübingen, Germany; University Hospital Tübingen, Internal Medicine IV, Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Otfried-Müller-Str. 10, 72076 Tübingen, Germany.
| |
Collapse
|
110
|
Mugabo Y, Zhao S, Lamontagne J, Al-Mass A, Peyot ML, Corkey BE, Joly E, Madiraju SRM, Prentki M. Metabolic fate of glucose and candidate signaling and excess-fuel detoxification pathways in pancreatic β-cells. J Biol Chem 2017; 292:7407-7422. [PMID: 28280244 DOI: 10.1074/jbc.m116.763060] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 03/06/2017] [Indexed: 12/28/2022] Open
Abstract
Glucose metabolism promotes insulin secretion in β-cells via metabolic coupling factors that are incompletely defined. Moreover, chronically elevated glucose causes β-cell dysfunction, but little is known about how cells handle excess fuels to avoid toxicity. Here we sought to determine which among the candidate pathways and coupling factors best correlates with glucose-stimulated insulin secretion (GSIS), define the fate of glucose in the β-cell, and identify pathways possibly involved in excess-fuel detoxification. We exposed isolated rat islets for 1 h to increasing glucose concentrations and measured various pathways and metabolites. Glucose oxidation, oxygen consumption, and ATP production correlated well with GSIS and saturated at 16 mm glucose. However, glucose utilization, glycerol release, triglyceride and glycogen contents, free fatty acid (FFA) content and release, and cholesterol and cholesterol esters increased linearly up to 25 mm glucose. Besides being oxidized, glucose was mainly metabolized via glycerol production and release and lipid synthesis (particularly FFA, triglycerides, and cholesterol), whereas glycogen production was comparatively low. Using targeted metabolomics in INS-1(832/13) cells, we found that several metabolites correlated well with GSIS, in particular some Krebs cycle intermediates, malonyl-CoA, and lower ADP levels. Glucose dose-dependently increased the dihydroxyacetone phosphate/glycerol 3-phosphate ratio in INS-1(832/13) cells, indicating a more oxidized state of NAD in the cytosol upon glucose stimulation. Overall, the data support a role for accelerated oxidative mitochondrial metabolism, anaplerosis, and malonyl-CoA/lipid signaling in β-cell metabolic signaling and suggest that a decrease in ADP levels is important in GSIS. The results also suggest that excess-fuel detoxification pathways in β-cells possibly comprise glycerol and FFA formation and release extracellularly and the diversion of glucose carbons to triglycerides and cholesterol esters.
Collapse
Affiliation(s)
- Yves Mugabo
- From the Montreal Diabetes Research Center and Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec H2X 0A9, Canada.,Departments of Nutrition, Biochemistry and Molecular Medicine, Faculty of Medicine, University of Montréal, Montreal, Québec H3C 3J7, Canada, and
| | - Shangang Zhao
- From the Montreal Diabetes Research Center and Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec H2X 0A9, Canada.,Departments of Medicine and Human Genetics, McGill University, Montreal, Québec H3A 1B1, Canada
| | - Julien Lamontagne
- From the Montreal Diabetes Research Center and Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec H2X 0A9, Canada
| | - Anfal Al-Mass
- From the Montreal Diabetes Research Center and Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec H2X 0A9, Canada.,Departments of Medicine and Human Genetics, McGill University, Montreal, Québec H3A 1B1, Canada
| | - Marie-Line Peyot
- From the Montreal Diabetes Research Center and Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec H2X 0A9, Canada
| | - Barbara E Corkey
- Department of Medicine, Obesity Research Center, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Erik Joly
- From the Montreal Diabetes Research Center and Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec H2X 0A9, Canada
| | - S R Murthy Madiraju
- From the Montreal Diabetes Research Center and Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec H2X 0A9, Canada
| | - Marc Prentki
- From the Montreal Diabetes Research Center and Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec H2X 0A9, Canada, .,Departments of Nutrition, Biochemistry and Molecular Medicine, Faculty of Medicine, University of Montréal, Montreal, Québec H3C 3J7, Canada, and
| |
Collapse
|
111
|
Barbarash O, Gruzdeva O, Uchasova E, Belik E, Dyleva Y, Karetnikova V. Biochemical markers of type 2 diabetes as a late complication of myocardial infarction: a case-control study. Arch Med Sci 2017; 13:311-320. [PMID: 28261283 PMCID: PMC5332457 DOI: 10.5114/aoms.2017.65240] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 02/25/2015] [Indexed: 01/17/2023] Open
Abstract
INTRODUCTION On average, 19-23% of patients with acute myocardial infarction (MI) suffer from type 2 diabetes mellitus, which is newly diagnosed in a significant number of patients. Both classic carbohydrate metabolism and lipid metabolism may be promising diagnostic markers for insulin resistance in acute coronary syndrome. MATERIAL AND METHODS Two hundred patients (130 males and 70 females aged 61.4 ±1.12 years) with ST-segment elevation MI were included in the study. Patients were divided into two groups based on manifestations of diabetes: (1) 171 patients without diabetes within 1 year after MI; and (2) 29 patients with manifestations of diabetes. The control group comprised 33 people without diseases of the cardiovascular system and diabetes and was matched by age and gender with patients. RESULTS In patients with an imbalanced adipokine state during the acute phase of MI, we noted an increased concentration of free fatty acids (p > 0.05) and reduced ghrelin levels (p > 0.05) and activation of the proinflammatory and thrombotic potentials of blood plasma. Patients who developed diabetes 1 year after MI showed hospital stays with more pronounced changes in the study parameters. CONCLUSIONS The most informative biochemical parameters associated with the development of diabetes at 1 year after MI were adiponectin, retinol protein, ghrelin, tumor necrosis factor α, and plasminogen activator inhibitor.
Collapse
Affiliation(s)
- Olga Barbarash
- Federal State Budgetary Scientific Institution Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - Olga Gruzdeva
- Federal State Budgetary Scientific Institution Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - Evgenya Uchasova
- Federal State Budgetary Scientific Institution Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - Ekaterina Belik
- Federal State Budgetary Scientific Institution Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - Yulia Dyleva
- Federal State Budgetary Scientific Institution Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - Victoria Karetnikova
- Federal State Budgetary Scientific Institution Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| |
Collapse
|
112
|
The Effects of Solvent Polarity on Hypoglycemic and Hypolipidemic Activities of Vaccinium Arctostaphylos L. Unripe Fruits. Pharm Chem J 2017. [DOI: 10.1007/s11094-017-1524-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
113
|
Conde-Sieira M, Soengas JL. Nutrient Sensing Systems in Fish: Impact on Food Intake Regulation and Energy Homeostasis. Front Neurosci 2017; 10:603. [PMID: 28111540 PMCID: PMC5216673 DOI: 10.3389/fnins.2016.00603] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 12/19/2016] [Indexed: 12/27/2022] Open
Abstract
Evidence obtained in recent years in a few species, especially rainbow trout, supports the presence in fish of nutrient sensing mechanisms. Glucosensing capacity is present in central (hypothalamus and hindbrain) and peripheral [liver, Brockmann bodies (BB, main accumulation of pancreatic endocrine cells in several fish species), and intestine] locations whereas fatty acid sensors seem to be present in hypothalamus, liver and BB. Glucose and fatty acid sensing capacities relate to food intake regulation and metabolism in fish. Hypothalamus is as a signaling integratory center in a way that detection of increased levels of nutrients result in food intake inhibition through changes in the expression of anorexigenic and orexigenic neuropeptides. Moreover, central nutrient sensing modulates functions in the periphery since they elicit changes in hepatic metabolism as well as in hormone secretion to counter-regulate changes in nutrient levels detected in the CNS. At peripheral level, the direct nutrient detection in liver has a crucial role in homeostatic control of glucose and fatty acid whereas in BB and intestine nutrient sensing is probably involved in regulation of hormone secretion from endocrine cells.
Collapse
Affiliation(s)
- Marta Conde-Sieira
- Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Universidade de Vigo Vigo, Spain
| | - José L Soengas
- Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Universidade de Vigo Vigo, Spain
| |
Collapse
|
114
|
Han T, Cheng Y, Tian S, Wang L, Liang X, Duan W, Na L, Sun C. Changes in triglycerides and high-density lipoprotein cholesterol may precede peripheral insulin resistance, with 2-h insulin partially mediating this unidirectional relationship: a prospective cohort study. Cardiovasc Diabetol 2016; 15:154. [PMID: 27814764 PMCID: PMC5095985 DOI: 10.1186/s12933-016-0469-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 10/26/2016] [Indexed: 12/31/2022] Open
Abstract
Background Results of longitudinal researches regarding the temporal relationship between dyslipidemia and insulin resistance (IR) are inconsistent. This study assessed temporal relationships of blood lipids with IR and determined whether there are any mediating effects existed in these temporal relationships. Methods This study examined a longitudinal cohort of 3325 subjects aged 20–74 years from China with an average of 4.2 years follow-up. Measurements of fasting blood lipids, as well as fasting and 2-h serum glucose and insulin, were obtained at two time points. The Gutt index and HOMA-IR were calculated as indicators of peripheral IR and hepatic IR. A cross-lagged path analysis was performed to examine the temporal relationships between blood lipids and IR. A mediation analysis was used to examine mediating effect. Results After adjusting for covariates, the cross-lagged path coefficients from baseline TG and HDL-C to follow-up Gutt index were significantly greater than those from baseline Gutt index to follow-up TG and HDL-C (β1 = −0.131 vs β2 = −0.047, P < 0.001 for TG; β1 = 0.134 vs β2 = 0.023, P < 0.001 for HDL-C). The path coefficients from baseline TG and HDL-C to follow-up 2-h insulin were significantly greater than those from baseline 2-h insulin to follow-up TG and HDL-C (β1 = 0.125 vs β2 = 0.040, P < 0.001 for TG; β1 = −0.112 vs β2 = −0.026, P < 0.001 for HDL-C). 2-h insulin partially mediated the effect of TG/HDL-C on Gutt index with a 59.3% mediating effect for TG and 61.0% for HDL-C. Conclusions These findings provide strong evidence that dyslipidemia probably precede peripheral IR and that 2-h insulin partially mediates this unidirectional temporal relationship. Electronic supplementary material The online version of this article (doi:10.1186/s12933-016-0469-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tianshu Han
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, 157 Baojian Road, Harbin, 150081, People's Republic of China
| | - Yu Cheng
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, 157 Baojian Road, Harbin, 150081, People's Republic of China
| | - Shuang Tian
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, 157 Baojian Road, Harbin, 150081, People's Republic of China
| | - Li Wang
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, 157 Baojian Road, Harbin, 150081, People's Republic of China
| | - Xi Liang
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, 157 Baojian Road, Harbin, 150081, People's Republic of China
| | - Wei Duan
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, 157 Baojian Road, Harbin, 150081, People's Republic of China
| | - Lixin Na
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, 157 Baojian Road, Harbin, 150081, People's Republic of China.
| | - Changhao Sun
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, 157 Baojian Road, Harbin, 150081, People's Republic of China.
| |
Collapse
|
115
|
Natalia A, Kim SJ, Kim HK. Antioxidant and antibacterial activity of fatty acid vanillyl ester produced by Proteus vulgaris K80 lipase-mediated transesterification. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/j.molcatb.2017.03.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
116
|
Kakei M, Yoshida M, Dezaki K, Ito K, Yamada H, Funazaki S, Kawakami M, Sugawara H, Yada T. Glucose and GTP-binding protein-coupled receptor cooperatively regulate transient receptor potential-channels to stimulate insulin secretion [Review]. Endocr J 2016; 63:867-876. [PMID: 27321586 DOI: 10.1507/endocrj.ej16-0262] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
In pancreatic β-cells, glucose-induced closure of the ATP-sensitive K+ (KATP) channel is an initial process triggering glucose-stimulated insulin secretion (GSIS). This KATP-channel dependent pathway has been believed to be a central mechanism for GSIS. However, since the resting membrane potential of cells is determined by the balance of the net result of current amplitudes in outward and inward directions, it must be taken into consideration that not only KATP channel inhibition but also inward current via the basal opening of non-selective cation channels (NSCCs) plays a crucial role in membrane potential regulation. The basal activity of NSCCs is essential to effectively evoke depolarization in concert with KATP channel closure that is dependent on glucose metabolism. The present study summarizes recent findings regarding the roles of NSCCs in GSIS and GTP-binding protein coupled receptor-(GPCR) operated potentiation of GSIS.
Collapse
Affiliation(s)
- Masafumi Kakei
- Internal Medicine, Saitama Medical Center, Jichi Medical University, Saitama 330-8503, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
117
|
Caporarello N, Salmeri M, Scalia M, Motta C, Parrino C, Frittitta L, Olivieri M, Cristaldi M, Avola R, Bramanti V, Toscano MA, Anfuso CD, Lupo G. Cytosolic and Calcium-Independent Phospholipases A2 Activation and Prostaglandins E2 Are Associated with Escherichia coli-Induced Reduction of Insulin Secretion in INS-1E Cells. PLoS One 2016; 11:e0159874. [PMID: 27631977 PMCID: PMC5024995 DOI: 10.1371/journal.pone.0159874] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 09/02/2016] [Indexed: 01/01/2023] Open
Abstract
It is suspected that microbial infections take part in the pathogenesis of diabetes mellitus type 1 (T1DM). Glucose-induced insulin secretion is accompanied by the release of free arachidonic acid (AA) mainly by cytosolic- and calcium independent phospholipases A2 (cPLA2 and iPLA2). Insulinoma cell line (INS-1E) was infected with E. coli isolated from the blood culture of a patient with sepsis. Invasion assay, Scanning Electron Microscopy and Transmission Electron Microscopy demonstrated the capacity of E. coli to enter cells, which was reduced by PLA2 inhibitors. Glucose-induced insulin secretion was significantly increased after acute infection (8h) but significantly decreased after chronic infection (72h). PLA2 activities, cPLA2, iPLA2, phospho-cPLA2, and COX-2 expressions were increased after acute and, even more, after chronic E. coli infection. The silencing of the two isoforms of PLA2s, with specific cPLA2- or iPLA2-siRNAs, reduced insulin secretion after acute infection and determined a rise in insulin release after chronic infection. Prostaglandins E2 (PGE2) production was significantly elevated in INS-1E after long-term E. coli infection and the restored insulin secretion in presence of L798106, a specific EP3 antagonist, and NS-398, a COX-2 inhibitor, and the reduction of insulin secretion in presence of sulprostone, a specific EP3 agonist, revealed their involvement in the effects triggered by bacterial infection. The results obtained demonstrated that cPLA2 and iPLA2 play a key role in insulin secretion process after E. coli infection. The high concentration of AA released is transformed into PGE2, which could be responsible for the reduced insulin secretion.
Collapse
Affiliation(s)
- Nunzia Caporarello
- Dept. of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania, Italy
| | - Mario Salmeri
- Dept. of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania, Italy
| | - Marina Scalia
- Dept. of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania, Italy
| | - Carla Motta
- Dept. of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania, Italy
| | - Cristina Parrino
- Dept. of Clinical and Experimental Medicine, School of Medicine, University of Catania, Catania, Italy
| | - Lucia Frittitta
- Dept. of Clinical and Experimental Medicine, School of Medicine, University of Catania, Catania, Italy
| | - Melania Olivieri
- Dept. of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania, Italy
| | - Martina Cristaldi
- Dept. of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania, Italy
| | - Roberto Avola
- Dept. of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania, Italy
| | - Vincenzo Bramanti
- Dept. of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania, Italy
| | - Maria Antonietta Toscano
- Dept. of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania, Italy
| | - Carmelina Daniela Anfuso
- Dept. of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania, Italy
| | - Gabriella Lupo
- Dept. of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania, Italy
- * E-mail:
| |
Collapse
|
118
|
Chen C, Chmelova H, Cohrs CM, Chouinard JA, Jahn SR, Stertmann J, Uphues I, Speier S. Alterations in β-Cell Calcium Dynamics and Efficacy Outweigh Islet Mass Adaptation in Compensation of Insulin Resistance and Prediabetes Onset. Diabetes 2016; 65:2676-85. [PMID: 27207518 DOI: 10.2337/db15-1718] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 04/05/2016] [Indexed: 11/13/2022]
Abstract
Emerging insulin resistance is normally compensated by increased insulin production of pancreatic β-cells, thereby maintaining normoglycemia. However, it is unclear whether this is achieved by adaptation of β-cell function, mass, or both. Most importantly, it is still unknown which of these adaptive mechanisms fail when type 2 diabetes develops. We performed longitudinal in vivo imaging of β-cell calcium dynamics and islet mass of transplanted islets of Langerhans throughout diet-induced progression from normal glucose homeostasis, through compensation of insulin resistance, to prediabetes. The results show that compensation of insulin resistance is predominated by alterations of β-cell function, while islet mass only gradually expands. Hereby, functional adaptation is mediated by increased calcium efficacy, which involves Epac signaling. Prior to prediabetes, β-cell function displays decreased stimulated calcium dynamics, whereas islet mass continues to increase through prediabetes onset. Thus, our data reveal a predominant role of islet function with distinct contributions of triggering and amplifying pathway in the in vivo processes preceding diabetes onset. These findings support protection and recovery of β-cell function as primary goals for prevention and treatment of diabetes and provide insight into potential therapeutic targets.
Collapse
Affiliation(s)
- Chunguang Chen
- Paul Langerhans Institute Dresden, Helmholtz Center Munich, University Clinic Carl Gustav Carus, Technische Universität Dresden, Helmholtz Zentrum München, Neuherberg, Germany German Research Foundation-Center for Regenerative Therapies Dresden (CRTD), Faculty of Medicine, Technische Universität Dresden, Dresden, Germany German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Helena Chmelova
- Paul Langerhans Institute Dresden, Helmholtz Center Munich, University Clinic Carl Gustav Carus, Technische Universität Dresden, Helmholtz Zentrum München, Neuherberg, Germany German Research Foundation-Center for Regenerative Therapies Dresden (CRTD), Faculty of Medicine, Technische Universität Dresden, Dresden, Germany German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Christian M Cohrs
- Paul Langerhans Institute Dresden, Helmholtz Center Munich, University Clinic Carl Gustav Carus, Technische Universität Dresden, Helmholtz Zentrum München, Neuherberg, Germany German Research Foundation-Center for Regenerative Therapies Dresden (CRTD), Faculty of Medicine, Technische Universität Dresden, Dresden, Germany German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Julie A Chouinard
- Paul Langerhans Institute Dresden, Helmholtz Center Munich, University Clinic Carl Gustav Carus, Technische Universität Dresden, Helmholtz Zentrum München, Neuherberg, Germany German Research Foundation-Center for Regenerative Therapies Dresden (CRTD), Faculty of Medicine, Technische Universität Dresden, Dresden, Germany German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Stephan R Jahn
- Paul Langerhans Institute Dresden, Helmholtz Center Munich, University Clinic Carl Gustav Carus, Technische Universität Dresden, Helmholtz Zentrum München, Neuherberg, Germany German Research Foundation-Center for Regenerative Therapies Dresden (CRTD), Faculty of Medicine, Technische Universität Dresden, Dresden, Germany German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Julia Stertmann
- Paul Langerhans Institute Dresden, Helmholtz Center Munich, University Clinic Carl Gustav Carus, Technische Universität Dresden, Helmholtz Zentrum München, Neuherberg, Germany German Research Foundation-Center for Regenerative Therapies Dresden (CRTD), Faculty of Medicine, Technische Universität Dresden, Dresden, Germany German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Ingo Uphues
- Boehringer Ingelheim Pharma GmbH & Co. KG, Ingelheim, Germany
| | - Stephan Speier
- Paul Langerhans Institute Dresden, Helmholtz Center Munich, University Clinic Carl Gustav Carus, Technische Universität Dresden, Helmholtz Zentrum München, Neuherberg, Germany German Research Foundation-Center for Regenerative Therapies Dresden (CRTD), Faculty of Medicine, Technische Universität Dresden, Dresden, Germany German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| |
Collapse
|
119
|
Frank JA, Yushchenko DA, Hodson DJ, Lipstein N, Nagpal J, Rutter GA, Rhee JS, Gottschalk A, Brose N, Schultz C, Trauner D. Photoswitchable diacylglycerols enable optical control of protein kinase C. Nat Chem Biol 2016; 12:755-62. [PMID: 27454932 PMCID: PMC6101201 DOI: 10.1038/nchembio.2141] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 05/16/2016] [Indexed: 01/02/2023]
Abstract
Increased levels of the second messenger lipid diacylglycerol (DAG) induce downstream signaling events including the translocation of C1-domain-containing proteins toward the plasma membrane. Here, we introduce three light-sensitive DAGs, termed PhoDAGs, which feature a photoswitchable acyl chain. The PhoDAGs are inactive in the dark and promote the translocation of proteins that feature C1 domains toward the plasma membrane upon a flash of UV-A light. This effect is quickly reversed after the termination of photostimulation or by irradiation with blue light, permitting the generation of oscillation patterns. Both protein kinase C and Munc13 can thus be put under optical control. PhoDAGs control vesicle release in excitable cells, such as mouse pancreatic islets and hippocampal neurons, and modulate synaptic transmission in Caenorhabditis elegans. As such, the PhoDAGs afford an unprecedented degree of spatiotemporal control and are broadly applicable tools to study DAG signaling.
Collapse
Affiliation(s)
- James Allen Frank
- Department of Chemistry and Center for Integrated Protein Science, Ludwig Maximilians University Munich, Munich, Germany
| | - Dmytro A Yushchenko
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - David J Hodson
- Section of Cell Biology and Functional Genomics, Department of Medicine, Imperial College London, ICTEM, Hammersmith Hospital, London, UK
- Institute of Metabolism and Systems Research (IMSR) and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism (CEDAM), Birmingham Health Partners, Birmingham, UK
| | - Noa Lipstein
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Jatin Nagpal
- Buchmann Institute for Molecular Life Sciences, Goethe University, Frankfurt, Germany
- Institute of Biochemistry, Department for Biochemistry, Chemistry and Pharmacy, Goethe University, Frankfurt, Germany
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Department of Medicine, Imperial College London, ICTEM, Hammersmith Hospital, London, UK
| | - Jeong-Seop Rhee
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Alexander Gottschalk
- Buchmann Institute for Molecular Life Sciences, Goethe University, Frankfurt, Germany
- Institute of Biochemistry, Department for Biochemistry, Chemistry and Pharmacy, Goethe University, Frankfurt, Germany
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Carsten Schultz
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Dirk Trauner
- Department of Chemistry and Center for Integrated Protein Science, Ludwig Maximilians University Munich, Munich, Germany
| |
Collapse
|
120
|
Moon JS, Nakahira K, Chung KP, DeNicola GM, Koo MJ, Pabón MA, Rooney KT, Yoon JH, Ryter SW, Stout-Delgado H, Choi AMK. NOX4-dependent fatty acid oxidation promotes NLRP3 inflammasome activation in macrophages. Nat Med 2016; 22:1002-12. [PMID: 27455510 PMCID: PMC5204248 DOI: 10.1038/nm.4153] [Citation(s) in RCA: 256] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 06/22/2016] [Indexed: 12/11/2022]
Abstract
Altered metabolism has been implicated in the pathogenesis of inflammatory diseases. NADPH oxidase 4 (NOX4), a source of cellular superoxide anions, has multiple biological functions that may be of importance in inflammation and in the pathogenesis of human metabolic diseases, including diabetes. However, the mechanisms by which NOX4-dependent metabolic regulation affect the innate immune response remain unclear. Here we show that deficiency of NOX4 resulted in reduced expression of carnitine palmitoyltransferase 1A (CPT1A), which is a key mitochondrial enzyme in the fatty acid oxidation (FAO) pathway. The reduced FAO resulted in less activation of the nucleotide-binding domain, leucine-rich-repeat-containing receptor (NLR), pyrin-domain-containing 3 (NLRP3) inflammasome in human and mouse macrophages. In contrast, NOX4 deficiency did not inhibit the activation of the NLR family, CARD-domain-containing 4 (NLRC4), the NLRP1 or the absent in melanoma 2 (AIM2) inflammasomes. We also found that inhibition of FAO by etomoxir treatment suppressed NLRP3 inflammasome activation. Furthermore, Nox4-deficient mice showed substantial reduction in caspase-1 activation and in interleukin (IL)-1β and IL-18 production, and there was improved survival in a mouse model of NLRP3-mediated Streptococcus pneumoniae infection. The pharmacologic inhibition of NOX4 by either GKT137831, which is currently in phase 2 clinical trials, or VAS-2870 attenuated NLRP3 inflammasome activation. Our results suggest that NOX4-mediated FAO promotes NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Jong-Seok Moon
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, USA
- Division of Pulmonary and Critical Care Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Kiichi Nakahira
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, USA
- Division of Pulmonary and Critical Care Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Kuei-Pin Chung
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, USA
- Division of Pulmonary and Critical Care Medicine, Weill Cornell Medical College, New York, NY, USA
- Department of Laboratory Medicine, National Taiwan University Hospital and National Taiwan University Cancer Center, Taipei, Taiwan
| | - Gina M. DeNicola
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, USA
| | - Michael Jakun Koo
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, USA
- Division of Pulmonary and Critical Care Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Maria A. Pabón
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, USA
| | - Kristen T. Rooney
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, USA
- Division of Pulmonary and Critical Care Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Joo-Heon Yoon
- Research Center for Natural Human Defense System, Yonsei University College of Medicine, Seoul, South Korea
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, South Korea
- The Airway Mucus Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Stefan W. Ryter
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, USA
- Division of Pulmonary and Critical Care Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Heather Stout-Delgado
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, USA
- Division of Pulmonary and Critical Care Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Augustine M. K. Choi
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, USA
- Division of Pulmonary and Critical Care Medicine, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
121
|
Salameh TS, Rhea EM, Banks WA, Hanson AJ. Insulin resistance, dyslipidemia, and apolipoprotein E interactions as mechanisms in cognitive impairment and Alzheimer's disease. Exp Biol Med (Maywood) 2016; 241:1676-83. [PMID: 27470930 PMCID: PMC4999626 DOI: 10.1177/1535370216660770] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
An increased risk for Alzheimer's disease is associated with dyslipidemia and insulin resistance. A separate literature shows the genetic risk for developing Alzheimer's disease is strongly correlated to the presence of the E4 isoform of the apolipoprotein E carrier protein. Understanding how apolipoprotein E carrier protein, lipids, amyloid β peptides, glucose, central nervous system insulin, and peripheral insulin interact with one another in Alzheimer's disease is an area of increasing interest. Here, we will review the evidence relating apolipoprotein E carrier protein, lipids, and insulin action to Alzheimer's disease and Aβ peptides and then propose mechanisms as to how these factors might interact with one another to impair cognition and promote Alzheimer's disease.
Collapse
Affiliation(s)
- Therese S Salameh
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Elizabeth M Rhea
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - William A Banks
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Angela J Hanson
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| |
Collapse
|
122
|
Gu J, Wei Q, Zheng H, Meng X, Zhang J, Wang D. Exendin-4 Promotes Survival of Mouse Pancreatic β-Cell Line in Lipotoxic Conditions, through the Extracellular Signal-Related Kinase 1/2 Pathway. J Diabetes Res 2016; 2016:5294025. [PMID: 27656657 PMCID: PMC5021481 DOI: 10.1155/2016/5294025] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 06/11/2016] [Accepted: 07/11/2016] [Indexed: 12/25/2022] Open
Abstract
Type 2 diabetes is a heterogeneous disorder that develops as a result of relatively inappropriate insulin secretion and insulin resistance. Increased levels of free fatty acids (FFAs) are one of the important factors for the pathogenesis of type 2 diabetes and contribute to defective β-cell proliferation and increased β-cell apoptosis. Recently, glucagon-like peptide-1 (GLP-1) receptor agonists have been shown to possess an antiapoptotic effect, by increasing β-cell mass and improving β-cell function. However, their effects on β-cells in vitro against lipotoxicity have not been elucidated completely. In this study, we investigated whether the GLP-1 receptor agonist exendin-4 displays prosurvival effects in pancreatic β-cells exposed to chronic elevated FFAs. Results showed that exendin-4 inhibited apoptosis induced by palmitate in MIN6 cells. After 24 h of incubation, exendin-4 caused rapid activation of extracellular signal-related kinase 1/2 (ERK1/2) under lipotoxic conditions. The ERK1/2 inhibitor PD98059 blocked the antilipotoxic effect of exendin-4 on MIN6 cells. Exendin-4 also inhibited the mitochondrial pathway of apoptosis. This inhibition is associated with upregulation of BCL-2. Our findings suggested that exendin-4 may exert cytoprotective effects through activation of ERK1/2 and inhibition of the mitochondrial apoptosis pathway.
Collapse
Affiliation(s)
- Jianqiu Gu
- Department of Endocrinology, The First Affiliated Hospital of China Medical University, 155 Nanjingbei Street, Heping District, Shenyang, Liaoning 110001, China
| | - Qian Wei
- Department of Geriatric Endocrinology, The First Affiliated Hospital of China Medical University, 155 Nanjingbei Street, Heping District, Shenyang, Liaoning 110001, China
| | - Hongzhi Zheng
- Department of Geriatric Endocrinology, The First Affiliated Hospital of China Medical University, 155 Nanjingbei Street, Heping District, Shenyang, Liaoning 110001, China
| | - Xin Meng
- Department of Endocrinology, The First Affiliated Hospital of China Medical University, 155 Nanjingbei Street, Heping District, Shenyang, Liaoning 110001, China
| | - Jin Zhang
- Department of Endocrinology, The First Affiliated Hospital of China Medical University, 155 Nanjingbei Street, Heping District, Shenyang, Liaoning 110001, China
| | - Difei Wang
- Department of Geriatric Endocrinology, The First Affiliated Hospital of China Medical University, 155 Nanjingbei Street, Heping District, Shenyang, Liaoning 110001, China
| |
Collapse
|
123
|
Molven A, Hollister-Lock J, Hu J, Martinez R, Njølstad PR, Liew CW, Weir G, Kulkarni RN. The Hypoglycemic Phenotype Is Islet Cell-Autonomous in Short-Chain Hydroxyacyl-CoA Dehydrogenase-Deficient Mice. Diabetes 2016; 65:1672-8. [PMID: 26953163 PMCID: PMC4878426 DOI: 10.2337/db15-1475] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 03/04/2016] [Indexed: 11/30/2022]
Abstract
Congenital hyperinsulinism of infancy (CHI) can be caused by inactivating mutations in the gene encoding short-chain 3-hydroxyacyl-CoA dehydrogenase (SCHAD), a ubiquitously expressed enzyme involved in fatty acid oxidation. The hypersecretion of insulin may be explained by a loss of interaction between SCHAD and glutamate dehydrogenase in the pancreatic β-cells. However, there is also a general accumulation of metabolites specific for the enzymatic defect in affected individuals. It remains to be explored whether hypoglycemia in SCHAD CHI can be uncoupled from the systemic effect on fatty acid oxidation. We therefore transplanted islets from global SCHAD knockout (SCHADKO) mice into mice with streptozotocin-induced diabetes. After transplantation, SCHADKO islet recipients exhibited significantly lower random and fasting blood glucose compared with mice transplanted with normal islets or nondiabetic, nontransplanted controls. Furthermore, intraperitoneal glucose tolerance was improved in animals receiving SCHADKO islets compared with those receiving normal islets. Graft β-cell proliferation and apoptosis rates were similar in the two transplantation groups. We conclude that hypoglycemia in SCHAD-CHI is islet cell-autonomous.
Collapse
Affiliation(s)
- Anders Molven
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, Bergen, Norway KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Jennifer Hollister-Lock
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA
| | - Jiang Hu
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA
| | - Rachael Martinez
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA
| | - Pål R Njølstad
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Chong Wee Liew
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL
| | - Gordon Weir
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA
| | - Rohit N Kulkarni
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA Harvard Stem Cell Institute, Boston, MA
| |
Collapse
|
124
|
Sun P, Wang T, Chen L, Yu BW, Jia Q, Chen KX, Fan HM, Li YM, Wang HY. Trimer procyanidin oligomers contribute to the protective effects of cinnamon extracts on pancreatic β-cells in vitro. Acta Pharmacol Sin 2016; 37:1083-90. [PMID: 27238208 DOI: 10.1038/aps.2016.29] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 03/28/2016] [Indexed: 11/09/2022] Open
Abstract
AIM Cinnamon extracts rich in procyanidin oligomers have shown to improve pancreatic β-cell function in diabetic db/db mice. The aim of this study was to identify the active compounds in extracts from two species of cinnamon responsible for the pancreatic β-cell protection in vitro. METHODS Cinnamon extracts were prepared from Cinnamomum tamala (CT-E) and Cinnamomum cassia (CC-E). Six compounds procyanidin B2 (cpd1), (-)-epicatechin (cpd2), cinnamtannin B1 (cpd3), procyanidin C1 (cpd4), parameritannin A1 (cpd5) and cinnamtannin D1 (cpd6) were isolated from the extracts. INS-1 pancreatic β-cells were exposed to palmitic acid (PA) or H2O2 to induce lipotoxicity and oxidative stress. Cell viability and apoptosis as well as ROS levels were assessed. Glucose-stimulated insulin secretion was examined in PA-treated β-cells and murine islets. RESULTS CT-E, CC-E as well as the compounds, except cpd5, did not cause cytotoxicity in the β-cells up to the maximum dosage using in this experiment. CT-E and CC-E (12.5-50 μg/mL) dose-dependently increased cell viability in both PA- and H2O2-treated β-cells, and decreased ROS accumulation in H2O2-treated β-cells. CT-E caused more prominent β-cell protection than CC-E. Furthermore, CT-E (25 and 50 μg/mL) dose-dependently increased glucose-stimulated insulin secretion in PA-treated β-cells and murine islets, but CC-E had little effect. Among the 6 compounds, trimer procyanidins cpd3, cpd4 and cpd6 (12.5-50 μmol/L) dose-dependently increased the cell viability and decreased ROS accumulation in H2O2-treated β-cells. The trimer procyanidins also increased glucose-stimulated insulin secretion in PA-treated β-cells. CONCLUSION Trimer procyanidins in the cinnamon extracts contribute to the pancreatic β-cell protection, thus to the anti-diabetic activity.
Collapse
Affiliation(s)
- Peng Sun
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ting Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Lu Chen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Bang-Wei Yu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Qi Jia
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Kai-Xian Chen
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hui-Min Fan
- Department of Cardiovascular and Thoracic Surgery, Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200123, China
| | - Yi-Ming Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - He-Yao Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
125
|
Li Z, Qiu Q, Geng X, Yang J, Huang W, Qian H. Free fatty acid receptor agonists for the treatment of type 2 diabetes: drugs in preclinical to phase II clinical development. Expert Opin Investig Drugs 2016; 25:871-90. [PMID: 27171154 DOI: 10.1080/13543784.2016.1189530] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION The alarming prevalence of type 2 diabetes mellitus (T2DM) stimulated the exploitation of new antidiabetic drugs with extended durability and enhanced safety. In this regard, the free fatty acid receptor 1 (FFA1) and FFA4 have emerged as attractive targets in the last decade. FFA1 has prominent advantages in promoting insulin and incretin secretion while FFA4 shows great potential in incretin secretion, insulin sensitization and anti-inflammatory effects. AREA COVERED Herein, the authors focus specifically on FFA1 and FFA4 agonists in clinical trials and preclinical development. LY2922470, P11187 and SHR0534 are currently active in clinical trials while the CNX-011-67, SAR1, DS-1558 and BMS-986118 are in preclinical phase. The information for this review is retrieved from Integrity, Scifinder, Espacenet and clinicaltrials.gov databases. EXPERT OPINION Current proof-of-concept in clinical trials suggests that FFA1 agonists have a significant improvement for T2DM without the risk of hypoglycemia. However, there are still several challenging problems including the mechanism of the receptor and the efficacy and safety of the ligands.
Collapse
Affiliation(s)
- Zheng Li
- a Center of Drug Discovery, State Key Laboratory of Natural Medicines , China Pharmaceutical University , Nanjing , PR China
| | - Qianqian Qiu
- a Center of Drug Discovery, State Key Laboratory of Natural Medicines , China Pharmaceutical University , Nanjing , PR China
| | - Xinqian Geng
- b Department of Endocrinology and Metabolism , Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes , Shanghai , PR China
| | - Jianyong Yang
- a Center of Drug Discovery, State Key Laboratory of Natural Medicines , China Pharmaceutical University , Nanjing , PR China
| | - Wenlong Huang
- a Center of Drug Discovery, State Key Laboratory of Natural Medicines , China Pharmaceutical University , Nanjing , PR China.,c Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease , China Pharmaceutical University , Nanjing , PR China
| | - Hai Qian
- a Center of Drug Discovery, State Key Laboratory of Natural Medicines , China Pharmaceutical University , Nanjing , PR China.,c Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease , China Pharmaceutical University , Nanjing , PR China
| |
Collapse
|
126
|
Yamada H, Yoshida M, Ito K, Dezaki K, Yada T, Ishikawa SE, Kakei M. Potentiation of Glucose-stimulated Insulin Secretion by the GPR40-PLC-TRPC Pathway in Pancreatic β-Cells. Sci Rep 2016; 6:25912. [PMID: 27180622 PMCID: PMC4867641 DOI: 10.1038/srep25912] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 04/25/2016] [Indexed: 01/04/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are expressed in pancreatic beta-cells. G protein-coupled receptor 40 (GPR40) contributes to medium- or long-chain fatty acid-induced amplification of glucose-stimulated insulin secretion (GSIS), and GPR40 agonists are promising therapeutic targets in type 2 diabetes. Recently, we demonstrated that glucagon-like peptide 1, a ligand of pancreatic GPCR, activates a class of nonselective cation channels (NSCCs) and enhances GSIS. The aim of the current study was to determine whether the GPR40 signal interacts with NSCCs. A GPR40 agonist (fasiglifam) potentiated GSIS at 8.3 and 16.7 mM glucose but not 2.8 mM glucose. The NSCC current was activated by fasiglifam at 5.6 mM glucose with 100 μM tolbutamide (−70 mV), and this activation was prevented by the presence of pyrazole-3 (transient receptor potential canonical; a TRPC3 channel blocker). Inhibitors of phospholipase C or protein kinase C (PKC) inhibited the increases in GSIS and the NSCC current induced by GPR40 stimulation. The present study demonstrates a novel mechanism for the regulation of insulin secretion by GPR40 agonist in pancreatic beta-cells. The stimulation of the GPR40–PLC/PKC–TRPC3 channel pathway potentiates GSIS by the depolarization of the plasma membrane in pancreatic beta-cell.
Collapse
Affiliation(s)
- Hodaka Yamada
- First Department of Comprehensive Medicine, Jichi Medical University Saitama Medical Center, Amanuma, Omiya 1-847, Saitama 330-8503, Japan
| | - Masashi Yoshida
- First Department of Comprehensive Medicine, Jichi Medical University Saitama Medical Center, Amanuma, Omiya 1-847, Saitama 330-8503, Japan
| | - Kiyonori Ito
- First Department of Comprehensive Medicine, Jichi Medical University Saitama Medical Center, Amanuma, Omiya 1-847, Saitama 330-8503, Japan
| | - Katsuya Dezaki
- Division of Integrative Physiology, Department of physiology, Jichi Medical University School of Medicine, Yakushiji 3311-1, Shimotsuke, Tochigi 329-0498, Japan
| | - Toshihiko Yada
- Division of Integrative Physiology, Department of physiology, Jichi Medical University School of Medicine, Yakushiji 3311-1, Shimotsuke, Tochigi 329-0498, Japan
| | - San-E Ishikawa
- First Department of Comprehensive Medicine, Jichi Medical University Saitama Medical Center, Amanuma, Omiya 1-847, Saitama 330-8503, Japan
| | - Masafumi Kakei
- First Department of Comprehensive Medicine, Jichi Medical University Saitama Medical Center, Amanuma, Omiya 1-847, Saitama 330-8503, Japan
| |
Collapse
|
127
|
Wang Y, Han C, Zhu W, Wu Z, Liu Y, Chen L. An optical method to evaluate both mass and functional competence of pancreatic α- and β-cells. J Cell Sci 2016; 129:2462-71. [PMID: 27173492 DOI: 10.1242/jcs.184523] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 04/28/2016] [Indexed: 01/09/2023] Open
Abstract
Imbalanced glucagon and insulin release leads to the onset of type 2 diabetes. To pinpoint the underlying primary driving force, here we have developed a fast, non-biased optical method to measure ratios of pancreatic α- and β-cell mass and function simultaneously. We firstly label both primary α- and β-cells with the red fluorescent probe ZinRhodaLactam-1 (ZRL1), and then highlight α-cells by selectively quenching the ZRL1 signal from β-cells. Based on the signals before and after quenching, we calculate the ratio of the α-cell to β-cell mass within live islets, which we found matched the results from immunohistochemistry. From the same islets, glucagon and insulin release capability can be concomitantly measured. Thus, we were able to measure the ratio of α-cell to β-cell mass and their function in wild-type and diabetic Lepr(db)/Lepr(db) (denoted db/db) mice at different ages. We find that the initial glucose intolerance that appears in 10-week-old db/db mice is associated with further expansion of α-cell mass prior to deterioration in functional β-cell mass. Our method is extendable to studies of islet mass and function in other type 2 diabetes animal models, which shall benefit mechanistic studies of imbalanced hormone secretion during type 2 diabetes progression.
Collapse
Affiliation(s)
- Yi Wang
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing 100871, China
| | - Chengsheng Han
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing 100871, China
| | - Wenzhen Zhu
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing 100871, China
| | - Zhengxing Wu
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yanmei Liu
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing 100871, China
| | - Liangyi Chen
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing 100871, China
| |
Collapse
|
128
|
Pearson GL, Mellett N, Chu KY, Boslem E, Meikle PJ, Biden TJ. A comprehensive lipidomic screen of pancreatic β-cells using mass spectroscopy defines novel features of glucose-stimulated turnover of neutral lipids, sphingolipids and plasmalogens. Mol Metab 2016; 5:404-414. [PMID: 27257600 PMCID: PMC4877660 DOI: 10.1016/j.molmet.2016.04.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 04/05/2016] [Accepted: 04/07/2016] [Indexed: 01/05/2023] Open
Abstract
Objective Glucose promotes lipid remodelling in pancreatic β-cells, and this is thought to contribute to the regulation of insulin secretion, but the metabolic pathways and potential signalling intermediates have not been fully elaborated. Methods Using mass spectrometry (MS) we quantified changes in approximately 300 lipid metabolites in MIN6 β-cells and isolated mouse islets following 1 h stimulation with glucose. Flux through sphingolipid pathways was also assessed in 3H-sphinganine-labelled cells using TLC. Results Glucose specifically activates the conversion of triacylglycerol (TAG) to diacylglycerol (DAG). This leads indirectly to the formation of 18:1 monoacylglycerol (MAG), via degradation of saturated/monounsaturated DAG species, such as 16:0_18:1 DAG, which are the most abundant, immediate products of glucose-stimulated TAG hydrolysis. However, 16:0-containing, di-saturated DAG species are a better direct marker of TAG hydrolysis since, unlike the 18:1-containing DAGs, they are predominately formed via this route. Using multiple reaction monitoring, we confirmed that in islets under basal conditions, 18:1 MAG is the most abundant species. We further demonstrated a novel site of glucose to enhance the conversion of ceramide to sphingomyelin (SM) and galactosylceramide (GalCer). Flux and product:precursor analyses suggest regulation of the enzyme SM synthase, which would constitute a separate mechanism for localized generation of DAG in response to glucose. Phosphatidylcholine (PC) plasmalogen (P) species, specifically those containing 20:4, 22:5 and 22:6 side chains, were also diminished in the presence of glucose, whereas the more abundant phosphatidylethanolamine plasmalogens were unchanged. Conclusion Our results highlight 18:1 MAG, GalCer, PC(P) and DAG/SM as potential contributors to metabolic stimulus-secretion coupling. Using mass spectroscopy lipidomics we have defined new aspects of glucose simulated lipid turnover in pancreatic beta cells. Glucose directly stimulates triacylglycerol hydrolysis, of which di-saturated diacylglycerol species are excellent markers. C18:1 is the most abundant monacylglycerol, and the one most obviously increased by glucose. Phosphatidylcholine plasmalogens with poly-unsaturated side chains are preferentially decreased by glucose. Glucose specifically enhances the conversion of ceramide to both sphingomyelin and galactosylceramide.
Collapse
Key Words
- (O), ether lipid
- (P), plasmalogen
- ATGL, adipose tissue glycerolipase
- CE, cholesterol ester
- COH, free cholesterol
- Ceramide
- DAG, diacylglycerol
- Diacylglycerol
- FA, fatty acid
- GSIS, glucose-stimulated insulin secretion
- GalCer, galactosylceramide
- GluCer, glucosylceramide
- HSL, hormone sensitive lipase
- Insulin secretion
- KRHB, Krebs Ringer Hepes Buffer
- MAG, monacylglycerol
- MHC, monohexosylceramide
- MS, mass spectrometry
- Monacylglycerol
- PC, phosphatidylcholine
- PE, phosphatidylethanolamine
- PI, phosphatidylinositol
- PKD, protein kinase D
- PLA2, phospholipase A2
- Pancreatic β-cell
- Plasmalogen
- SM, sphingomyelin
- TAG, triacylglycerol
Collapse
Affiliation(s)
- Gemma L Pearson
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW, 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Australia
| | - Natalie Mellett
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Kwan Yi Chu
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW, 2010, Australia
| | - Ebru Boslem
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW, 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Australia
| | - Peter J Meikle
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia.
| | - Trevor J Biden
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW, 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Australia.
| |
Collapse
|
129
|
Pepin É, Al-Mass A, Attané C, Zhang K, Lamontagne J, Lussier R, Madiraju SRM, Joly E, Ruderman NB, Sladek R, Prentki M, Peyot ML. Pancreatic β-Cell Dysfunction in Diet-Induced Obese Mice: Roles of AMP-Kinase, Protein Kinase Cε, Mitochondrial and Cholesterol Metabolism, and Alterations in Gene Expression. PLoS One 2016; 11:e0153017. [PMID: 27043434 PMCID: PMC4820227 DOI: 10.1371/journal.pone.0153017] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 03/22/2016] [Indexed: 12/27/2022] Open
Abstract
Diet induced obese (DIO) mice can be stratified according to their weight gain in response to high fat diet as low responders (LDR) and high responders (HDR). This allows the study of β-cell failure and the transitions to prediabetes (LDR) and early diabetes (HDR). C57BL/6N mice were fed for 8 weeks with a normal chow diet (ND) or a high fat diet and stratified as LDR and HDR. Freshly isolated islets from ND, LDR and HDR mice were studied ex-vivo for mitochondrial metabolism, AMPK activity and signalling, the expression and activity of key enzymes of energy metabolism, cholesterol synthesis, and mRNA profiling. Severely compromised glucose-induced insulin secretion in HDR islets, as compared to ND and LDR islets, was associated with suppressed AMP-kinase activity. HDR islets also showed reduced acetyl-CoA carboxylase activity and enhanced activity of 3-hydroxy-3-methylglutaryl-CoA reductase, which led respectively to elevated fatty acid oxidation and increased cholesterol biosynthesis. HDR islets also displayed mitochondrial membrane hyperpolarization and reduced ATP turnover in the presence of elevated glucose. Expression of protein kinase Cε, which reduces both lipolysis and production of signals for insulin secretion, was elevated in DIO islets. Genes whose expression increased or decreased by more than 1.2-fold were minor between LDR and ND islets (17 differentially expressed), but were prominent between HDR and ND islets (1508 differentially expressed). In HDR islets, particularly affected genes were related to cell cycle and proliferation, AMPK signaling, mitochondrial metabolism and cholesterol metabolism. In conclusion, chronically reduced AMPK activity, mitochondrial dysfunction, elevated cholesterol biosynthesis in islets, and substantial alterations in gene expression accompany β-cell failure in HDR islets. The β-cell compensation process in the prediabetic state (LDR) is largely independent of transcriptional adaptive changes, whereas the transition to early diabetes (HDR) is associated with major alterations in gene expression.
Collapse
Affiliation(s)
- Émilie Pepin
- Montreal Diabetes Research Center and Centre de Recherche du CHUM, Montréal, Québec, Canada
| | - Anfal Al-Mass
- Montreal Diabetes Research Center and Centre de Recherche du CHUM, Montréal, Québec, Canada
- Departments of Medicine and Human Genetics, McGill University, Montreal, Québec, Canada
| | - Camille Attané
- Montreal Diabetes Research Center and Centre de Recherche du CHUM, Montréal, Québec, Canada
| | - Kezhuo Zhang
- Departments of Medicine and Human Genetics, McGill University, Montreal, Québec, Canada
| | - Julien Lamontagne
- Montreal Diabetes Research Center and Centre de Recherche du CHUM, Montréal, Québec, Canada
| | - Roxane Lussier
- Montreal Diabetes Research Center and Centre de Recherche du CHUM, Montréal, Québec, Canada
| | - S. R. Murthy Madiraju
- Montreal Diabetes Research Center and Centre de Recherche du CHUM, Montréal, Québec, Canada
| | - Erik Joly
- Montreal Diabetes Research Center and Centre de Recherche du CHUM, Montréal, Québec, Canada
| | - Neil B. Ruderman
- Departments of Medicine and Physiology and Biophysics, Boston University School of Medicine and Diabetes Unit, Boston Medical Center, Boston, MA, United States of America
| | - Robert Sladek
- Departments of Medicine and Human Genetics, McGill University, Montreal, Québec, Canada
| | - Marc Prentki
- Montreal Diabetes Research Center and Centre de Recherche du CHUM, Montréal, Québec, Canada
- Departments of Nutrition, Biochemistry and Molecular Medicine, Faculty of Medicine, University of Montréal, Montreal, Québec, Canada
- * E-mail: (MP); (MLP)
| | - Marie-Line Peyot
- Montreal Diabetes Research Center and Centre de Recherche du CHUM, Montréal, Québec, Canada
- * E-mail: (MP); (MLP)
| |
Collapse
|
130
|
Polvani S, Tarocchi M, Tempesti S, Bencini L, Galli A. Peroxisome proliferator activated receptors at the crossroad of obesity, diabetes, and pancreatic cancer. World J Gastroenterol 2016; 22:2441-2459. [PMID: 26937133 PMCID: PMC4768191 DOI: 10.3748/wjg.v22.i8.2441] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 12/17/2015] [Accepted: 01/11/2016] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the fourth cause of cancer death with an overall survival of 5% at five years. The development of PDAC is characteristically associated to the accumulation of distinctive genetic mutations and is preceded by the exposure to several risk factors. Epidemiology has demonstrated that PDAC risk factors may be non-modifiable risks (sex, age, presence of genetic mutations, ethnicity) and modifiable and co-morbidity factors related to the specific habits and lifestyle. Recently it has become evident that obesity and diabetes are two important modifiable risk factors for PDAC. Obesity and diabetes are complex systemic and intertwined diseases and, over the years, experimental evidence indicate that insulin-resistance, alteration of adipokines, especially leptin and adiponectin, oxidative stress and inflammation may play a role in PDAC. Peroxisome proliferator activated receptor-γ (PPARγ) is a nuclear receptor transcription factor that is implicated in the regulation of metabolism, differentiation and inflammation. PPARγ is a key regulator of adipocytes differentiation, regulates insulin and adipokines production and secretion, may modulate inflammation, and it is implicated in PDAC. PPARγ agonists are used in the treatment of diabetes and oxidative stress-associated diseases and have been evaluated for the treatment of PDAC. PPARγ is at the cross-road of diabetes, obesity, and PDAC and it is an interesting target to pharmacologically prevent PDAC in obese and diabetic patients.
Collapse
|
131
|
Antidiabetic and Antilipidemic Effects of Some Polar and Nonpolar Extracts of Securigera Securidaca Flowers. Pharm Chem J 2016. [DOI: 10.1007/s11094-016-1365-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
132
|
Imbalanced insulin action in chronic over nutrition: Clinical harm, molecular mechanisms, and a way forward. Atherosclerosis 2016; 247:225-82. [PMID: 26967715 DOI: 10.1016/j.atherosclerosis.2016.02.004] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 12/31/2015] [Accepted: 02/02/2016] [Indexed: 02/08/2023]
Abstract
The growing worldwide prevalence of overnutrition and underexertion threatens the gains that we have made against atherosclerotic cardiovascular disease and other maladies. Chronic overnutrition causes the atherometabolic syndrome, which is a cluster of seemingly unrelated health problems characterized by increased abdominal girth and body-mass index, high fasting and postprandial concentrations of cholesterol- and triglyceride-rich apoB-lipoproteins (C-TRLs), low plasma HDL levels, impaired regulation of plasma glucose concentrations, hypertension, and a significant risk of developing overt type 2 diabetes mellitus (T2DM). In addition, individuals with this syndrome exhibit fatty liver, hypercoagulability, sympathetic overactivity, a gradually rising set-point for body adiposity, a substantially increased risk of atherosclerotic cardiovascular morbidity and mortality, and--crucially--hyperinsulinemia. Many lines of evidence indicate that each component of the atherometabolic syndrome arises, or is worsened by, pathway-selective insulin resistance and responsiveness (SEIRR). Individuals with SEIRR require compensatory hyperinsulinemia to control plasma glucose levels. The result is overdrive of those pathways that remain insulin-responsive, particularly ERK activation and hepatic de-novo lipogenesis (DNL), while carbohydrate regulation deteriorates. The effects are easily summarized: if hyperinsulinemia does something bad in a tissue or organ, that effect remains responsive in the atherometabolic syndrome and T2DM; and if hyperinsulinemia might do something good, that effect becomes resistant. It is a deadly imbalance in insulin action. From the standpoint of human health, it is the worst possible combination of effects. In this review, we discuss the origins of the atherometabolic syndrome in our historically unprecedented environment that only recently has become full of poorly satiating calories and incessant enticements to sit. Data are examined that indicate the magnitude of daily caloric imbalance that causes obesity. We also cover key aspects of healthy, balanced insulin action in liver, endothelium, brain, and elsewhere. Recent insights into the molecular basis and pathophysiologic harm from SEIRR in these organs are discussed. Importantly, a newly discovered oxide transport chain functions as the master regulator of the balance amongst different limbs of the insulin signaling cascade. This oxide transport chain--abbreviated 'NSAPP' after its five major proteins--fails to function properly during chronic overnutrition, resulting in this harmful pattern of SEIRR. We also review the origins of widespread, chronic overnutrition. Despite its apparent complexity, one factor stands out. A sophisticated junk food industry, aided by subsidies from willing governments, has devoted years of careful effort to promote overeating through the creation of a new class of food and drink that is low- or no-cost to the consumer, convenient, savory, calorically dense, yet weakly satiating. It is past time for the rest of us to overcome these foes of good health and solve this man-made epidemic.
Collapse
|
133
|
Shah M, Varghese RT, Miles JM, Piccinini F, Dalla Man C, Cobelli C, Bailey KR, Rizza RA, Vella A. TCF7L2 Genotype and α-Cell Function in Humans Without Diabetes. Diabetes 2016; 65:371-80. [PMID: 26525881 PMCID: PMC4747457 DOI: 10.2337/db15-1233] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 10/26/2015] [Indexed: 12/20/2022]
Abstract
The diabetes-associated allele in TCF7L2 increases the rate of conversion to diabetes; however, the mechanism by which this occurs remains elusive. We hypothesized that the diabetes-associated allele in this locus (rs7903146) impairs insulin secretion and that this defect would be exacerbated by acute free fatty acid (FFA)-induced insulin resistance. We studied 120 individuals of whom one-half were homozygous for the diabetes-associated allele TT at rs7903146 and one-half were homozygous for the protective allele CC. After a screening examination during which glucose tolerance status was determined, subjects were studied on two occasions in random order while undergoing an oral challenge. During one study day, FFA was elevated by infusion of Intralipid plus heparin. On the other study day, subjects received the same amount of glycerol as present in the Intralipid infusion. β-Cell responsivity indices were estimated with the oral C-peptide minimal model. We report that β-cell responsivity was slightly impaired in the TT genotype group. Moreover, the hyperbolic relationship between insulin secretion and β-cell responsivity differed significantly between genotypes. Subjects also exhibited impaired suppression of glucagon after an oral challenge. These data imply that a genetic variant harbored within the TCF7L2 locus impairs glucose tolerance through effects on glucagon as well as on insulin secretion.
Collapse
Affiliation(s)
- Meera Shah
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition Research, Mayo Clinic, Rochester, MN
| | - Ron T Varghese
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition Research, Mayo Clinic, Rochester, MN
| | - John M Miles
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition Research, Mayo Clinic, Rochester, MN
| | - Francesca Piccinini
- Department of Information Engineering, Università degli Studi di Padova, Padova, Italy
| | - Chiara Dalla Man
- Department of Information Engineering, Università degli Studi di Padova, Padova, Italy
| | - Claudio Cobelli
- Department of Information Engineering, Università degli Studi di Padova, Padova, Italy
| | - Kent R Bailey
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN
| | - Robert A Rizza
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition Research, Mayo Clinic, Rochester, MN
| | - Adrian Vella
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition Research, Mayo Clinic, Rochester, MN
| |
Collapse
|
134
|
Caporarello N, Salmeri M, Scalia M, Motta C, Parrino C, Frittitta L, Olivieri M, Toscano MA, Anfuso CD, Lupo G. Role of cytosolic and calcium independent phospholipases A(2) in insulin secretion impairment of INS-1E cells infected by S. aureus. FEBS Lett 2015; 589:3969-76. [PMID: 26632509 DOI: 10.1016/j.febslet.2015.11.035] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 11/19/2015] [Indexed: 10/22/2022]
Abstract
Cytosolic PLA2 (cPLA2) and Ca(2+)-independent PLA2 (iPLA2) play a significant role in insulin β-cells secretion. Bacterial infections may be responsible of the onset of diabetes. The mechanism by which Staphylococcus aureus infection of INS-1 cells alters glucose-induced insulin secretion has been examined. After acute infection, insulin secretion and PLA2 activities significantly increased. Moreover, increased expressions of phospho-cPLA2, phospho-PKCα and phospho-ERK 1/2 were observed. Chronic infection causes a decrease in insulin release and a significant increase of iPLA2 and COX-2 protein expression. Moreover, insulin secretion in infected cells could be restored using specific siRNAs against iPLA2 isoform and specific COX-2 inhibitor.
Collapse
Affiliation(s)
- N Caporarello
- Dept. of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Italy
| | - M Salmeri
- Dept. of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Italy
| | - M Scalia
- Dept. of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Italy
| | - C Motta
- Dept. of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Italy
| | - C Parrino
- Dept. of Clinical and Experimental Medicine, School of Medicine, University of Catania, Italy
| | - L Frittitta
- Dept. of Clinical and Experimental Medicine, School of Medicine, University of Catania, Italy
| | - M Olivieri
- Dept. of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Italy
| | - M A Toscano
- Dept. of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Italy
| | - C D Anfuso
- Dept. of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Italy
| | - G Lupo
- Dept. of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Italy.
| |
Collapse
|
135
|
|
136
|
Abstract
Insulin is a key hormone controlling metabolic homeostasis. Loss or dysfunction of pancreatic β-cells lead to the release of insufficient insulin to cover the organism needs, promoting diabetes development. Since dietary nutrients influence the activity of β-cells, their inadequate intake, absorption and/or utilisation can be detrimental. This review will highlight the physiological and pathological effects of nutrients on insulin secretion and discuss the underlying mechanisms. Glucose uptake and metabolism in β-cells trigger insulin secretion. This effect of glucose is potentiated by amino acids and fatty acids, as well as by entero-endocrine hormones and neuropeptides released by the digestive tract in response to nutrients. Glucose controls also basal and compensatory β-cell proliferation and, along with fatty acids, regulates insulin biosynthesis. If in the short-term nutrients promote β-cell activities, chronic exposure to nutrients can be detrimental to β-cells and causes reduced insulin transcription, increased basal secretion and impaired insulin release in response to stimulatory glucose concentrations, with a consequent increase in diabetes risk. Likewise, suboptimal early-life nutrition (e.g. parental high-fat or low-protein diet) causes altered β-cell mass and function in adulthood. The mechanisms mediating nutrient-induced β-cell dysfunction include transcriptional, post-transcriptional and translational modifications of genes involved in insulin biosynthesis and secretion, carbohydrate and lipid metabolism, cell differentiation, proliferation and survival. Altered expression of these genes is partly caused by changes in non-coding RNA transcripts induced by unbalanced nutrient uptake. A better understanding of the mechanisms leading to β-cell dysfunction will be critical to improve treatment and find a cure for diabetes.
Collapse
|
137
|
Moore WT, Bowser SM, Fausnacht DW, Staley LL, Suh KS, Liu D. Beta Cell Function and the Nutritional State: Dietary Factors that Influence Insulin Secretion. Curr Diab Rep 2015; 15:76. [PMID: 26294335 DOI: 10.1007/s11892-015-0650-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Approximately 366 million people worldwide have been diagnosed with type-2 diabetes (T2D). Chronic insulin resistance, decreased functional β-cell mass, and elevated blood glucose are defining characteristics of T2D. Great advances have been made in understanding the pathogenesis of T2D with respect to the effects of dietary macronutrient composition and energy intake on β-cell physiology and glucose homeostasis. It has been further established that obesity is a leading pathogenic factor for developing insulin resistance. However, insulin resistance may not progress to T2D unless β-cells are unable to secret an adequate amount of insulin to compensate for decreased insulin sensitivity. Therefore, pancreatic β-cell dysfunction plays an important role in the development of overt diabetes. This paper reviews recent research findings on the effects of several micronutrients (zinc, vitamin D, iron, vitamin A), leucine, and the phytochemical, genistein on pancreatic β-cell physiology with emphasis on their effects on insulin secretion, specifically in the context of T2D.
Collapse
Affiliation(s)
- William T Moore
- Department of Human Nutrition, Foods and Exercises, College of Agricultural and Life Sciences, Virginia Tech Corporate Research Center, 1981 Kraft Drive, Blacksburg, VA, 24061, USA
| | | | | | | | | | | |
Collapse
|
138
|
Cross-linking of sodium caseinate-structured emulsion with transglutaminase alters postprandial metabolic and appetite responses in healthy young individuals. Br J Nutr 2015; 114:418-29. [DOI: 10.1017/s0007114515001737] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The physico-chemical and interfacial properties of fat emulsions influence lipid digestion and may affect postprandial responses. The aim of the present study was to determine the effects of the modification of the interfacial layer of a fat emulsion by cross-linking on postprandial metabolic and appetite responses. A total of fifteen healthy individuals (26·5 (sem6·9) years and BMI 21·9 (sem2·0) kg/m2) participated in a cross-over design experiment in which they consumed two isoenergetic (1924 kJ (460 kcal)) and isovolumic (250 g) emulsions stabilised with either sodium caseinate (Cas) or transglutaminase-cross-linked sodium caseinate (Cas-TG) in a randomised order. Blood samples were collected from the individuals at baseline and for 6 h postprandially for the determination of serum TAG and plasma NEFA, cholecystokinin (CCK), glucagon-like peptide 1 (GLP-1), glucose and insulin responses. Appetite was assessed using visual analogue scales. Postprandial TAG and NEFA responses and gastric emptying (GE) rates were comparable between the emulsions. CCK increased more after the ingestion of Cas-TG than after the ingestion of Cas (P< 0·05), while GLP-1 responses did not differ between the two test emulsions. Glucose and insulin profiles were lower after consuming Cas-TG than after consuming Cas (P< 0·05). The overall insulin, glucose and CCK responses, expressed as areas above/under the curve, did not differ significantly between the Cas and Cas-TG meal conditions. Satiety ratings were reduced and hunger, desire to eat and thirst ratings increased more after the ingestion of Cas-TG than after the ingestion of Cas (P< 0·05). The present results suggest that even a subtle structural modification of the interfacial layer of a fat emulsion can alter the early postprandial profiles of glucose, insulin, CCK, appetite and satiety through decreased protein digestion without affecting significantly on GE or overall lipid digestion.
Collapse
|
139
|
Zhou Y, Sun P, Wang T, Chen K, Zhu W, Wang H. Inhibition of Calcium Influx Reduces Dysfunction and Apoptosis in Lipotoxic Pancreatic β-Cells via Regulation of Endoplasmic Reticulum Stress. PLoS One 2015; 10:e0132411. [PMID: 26147439 PMCID: PMC4492560 DOI: 10.1371/journal.pone.0132411] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 06/12/2015] [Indexed: 01/12/2023] Open
Abstract
Lipotoxicity plays an important role in pancreatic β-cell failure during the development of type 2 diabetes. Prolonged exposure of β-cells to elevated free fatty acids level could cause deterioration of β-cell function and induce cell apoptosis. Therefore, inhibition of fatty acids-induced β-cell dysfunction and apoptosis might provide benefit for the therapy of type 2 diabetes. The present study examined whether regulation of fatty acids-triggered calcium influx could protect pancreatic β-cells from lipotoxicity. Two small molecule compounds, L-type calcium channel blocker nifedipine and potassium channel activator diazoxide were used to inhibit palmitic acid-induced calcium influx. And whether the compounds could reduce palmitic acid-induced β-cell failure and the underlying mechanism were also investigated. It was found that both nifedipine and diazoxide protected MIN6 pancreatic β-cells and primary cultured murine islets from palmitic acid-induced apoptosis. Meanwhile, the impaired insulin secretion was also recovered to varying degrees by these two compounds. Our results verified that nifedipine and diazoxide could reduce palmitic acid-induced endoplasmic reticulum stress to generate protective effects on pancreatic β-cells. More importantly, it suggested that regulation of calcium influx by small molecule compounds might provide benefits for the prevention and therapy of type 2 diabetes.
Collapse
Affiliation(s)
- Yuren Zhou
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
| | - Peng Sun
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Ting Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
| | - Kaixian Chen
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Weiliang Zhu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
| | - Heyao Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
- * E-mail:
| |
Collapse
|
140
|
Metabolomics applied to the pancreatic islet. Arch Biochem Biophys 2015; 589:120-30. [PMID: 26116790 DOI: 10.1016/j.abb.2015.06.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 06/19/2015] [Accepted: 06/21/2015] [Indexed: 01/18/2023]
Abstract
Metabolomics, the characterization of the set of small molecules in a biological system, is advancing research in multiple areas of islet biology. Measuring a breadth of metabolites simultaneously provides a broad perspective on metabolic changes as the islets respond dynamically to metabolic fuels, hormones, or environmental stressors. As a result, metabolomics has the potential to provide new mechanistic insights into islet physiology and pathophysiology. Here we summarize advances in our understanding of islet physiology and the etiologies of type-1 and type-2 diabetes gained from metabolomics studies.
Collapse
|
141
|
Cohen G, Shamni O, Avrahami Y, Cohen O, Broner EC, Filippov-Levy N, Chatgilialoglu C, Ferreri C, Kaiser N, Sasson S. Beta cell response to nutrient overload involves phospholipid remodelling and lipid peroxidation. Diabetologia 2015; 58:1333-43. [PMID: 25810039 DOI: 10.1007/s00125-015-3566-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 03/11/2015] [Indexed: 12/13/2022]
Abstract
AIMS/HYPOTHESIS Membrane phospholipids are the major intracellular source for fatty acid-derived mediators, which regulate myriad cell functions. We showed previously that high glucose levels triggered the hydrolysis of polyunsaturated fatty acids from beta cell phospholipids. These fatty acids were subjected to free radical-catalysed peroxidation to generate the bioactive aldehyde 4-hydroxy-2E-nonenal (4-HNE). The latter activated the nuclear peroxisome proliferator-activated receptor-δ (PPARδ), which in turn augmented glucose-stimulated insulin secretion. The present study aimed at investigating the combined effects of glucose and fatty acid overload on phospholipid turnover and the subsequent generation of lipid mediators, which affect insulin secretion and beta cell viability. METHODS INS-1E cells were incubated with increasing glucose concentrations (5-25 mmol/l) without or with palmitic acid (PA; 50-500 μmol/l) and taken for fatty acid-based lipidomic analysis and functional assays. Rat isolated islets of Langerhans were used similarly. RESULTS PA was incorporated into membrane phospholipids in a concentration- and time-dependent manner; incorporation was highest at 25 mmol/l glucose. This was coupled to a rapid exchange with saturated, mono-unsaturated and polyunsaturated fatty acids. Importantly, released arachidonic acid and linoleic acid were subjected to peroxidation, resulting in the generation of 4-HNE, which further augmented insulin secretion by activating PPARδ in beta cells. However, this adaptive increase in insulin secretion was abolished at high glucose and PA levels, which induced endoplasmic reticulum stress, apoptosis and cell death. CONCLUSIONS/INTERPRETATION These findings highlight a key role for phospholipid remodelling and fatty acid peroxidation in mediating adaptive and cytotoxic interactions induced by nutrient overload in beta cells.
Collapse
Affiliation(s)
- Guy Cohen
- Department of Pharmacology, Institute for Drug Research, Faculty of Medicine, The Hebrew University, Jerusalem, 9112102, Israel
| | | | | | | | | | | | | | | | | | | |
Collapse
|
142
|
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
|
143
|
Erion KA, Berdan CA, Burritt NE, Corkey BE, Deeney JT. Chronic Exposure to Excess Nutrients Left-shifts the Concentration Dependence of Glucose-stimulated Insulin Secretion in Pancreatic β-Cells. J Biol Chem 2015; 290:16191-201. [PMID: 25934392 DOI: 10.1074/jbc.m114.620351] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Indexed: 01/21/2023] Open
Abstract
Hyperinsulinemia (HI) is elevated plasma insulin at basal glucose. Impaired glucose tolerance is associated with HI, although the exact cause and effect relationship remains poorly defined. We tested the hypothesis that HI can result from an intrinsic response of the β-cell to chronic exposure to excess nutrients, involving a shift in the concentration dependence of glucose-stimulated insulin secretion. INS-1 (832/13) cells were cultured in either a physiological (4 mm) or high (11 mm) glucose concentration with or without concomitant exposure to oleate. Isolated rat islets were also cultured with or without oleate. A clear hypersensitivity to submaximal glucose concentrations was evident in INS-1 cells cultured in excess nutrients such that the 25% of maximal (S0.25) glucose-stimulated insulin secretion was significantly reduced in cells cultured in 11 mm glucose (S0.25 = 3.5 mm) and 4 mm glucose with oleate (S0.25 = 4.5 mm) compared with 4 mm glucose alone (S0.25 = 5.7 mm). The magnitude of the left shift was linearly correlated with intracellular lipid stores in INS-1 cells (r(2) = 0.97). We observed no significant differences in the dose responses for glucose stimulation of respiration, NAD(P)H autofluorescence, or Ca(2+) responses between left- and right-shifted β-cells. However, a left shift in the sensitivity of exocytosis to Ca(2+) was documented in permeabilized INS-1 cells cultured in 11 versus 4 mm glucose (S0.25 = 1.1 and 1.7 μm, respectively). Our results suggest that the sensitivity of exocytosis to triggering is modulated by a lipid component, the levels of which are influenced by the culture nutrient environment.
Collapse
Affiliation(s)
- Karel A Erion
- From the Obesity Research Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Charles A Berdan
- From the Obesity Research Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Nathan E Burritt
- From the Obesity Research Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Barbara E Corkey
- From the Obesity Research Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Jude T Deeney
- From the Obesity Research Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118
| |
Collapse
|
144
|
Santulli G, Pagano G, Sardu C, Xie W, Reiken S, D'Ascia SL, Cannone M, Marziliano N, Trimarco B, Guise TA, Lacampagne A, Marks AR. Calcium release channel RyR2 regulates insulin release and glucose homeostasis. J Clin Invest 2015; 125:1968-78. [PMID: 25844899 DOI: 10.1172/jci79273] [Citation(s) in RCA: 152] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 02/27/2015] [Indexed: 12/21/2022] Open
Abstract
The type 2 ryanodine receptor (RyR2) is a Ca2+ release channel on the endoplasmic reticulum (ER) of several types of cells, including cardiomyocytes and pancreatic β cells. In cardiomyocytes, RyR2-dependent Ca2+ release is critical for excitation-contraction coupling; however, a functional role for RyR2 in β cell insulin secretion and diabetes mellitus remains controversial. Here, we took advantage of rare RyR2 mutations that were identified in patients with a genetic form of exercise-induced sudden death (catecholaminergic polymorphic ventricular tachycardia [CPVT]). As these mutations result in a "leaky" RyR2 channel, we exploited them to assess RyR2 channel function in β cell dynamics. We discovered that CPVT patients with mutant leaky RyR2 present with glucose intolerance, which was heretofore unappreciated. In mice, transgenic expression of CPVT-associated RyR2 resulted in impaired glucose homeostasis, and an in-depth evaluation of pancreatic islets and β cells from these animals revealed intracellular Ca2+ leak via oxidized and nitrosylated RyR2 channels, activated ER stress response, mitochondrial dysfunction, and decreased fuel-stimulated insulin release. Additionally, we verified the effects of the pharmacological inhibition of intracellular Ca2+ leak in CPVT-associated RyR2-expressing mice, in human islets from diabetic patients, and in an established murine model of type 2 diabetes mellitus. Taken together, our data indicate that RyR2 channels play a crucial role in the regulation of insulin secretion and glucose homeostasis.
Collapse
|
145
|
Wauquier F, Léotoing L, Philippe C, Spilmont M, Coxam V, Wittrant Y. Pros and cons of fatty acids in bone biology. Prog Lipid Res 2015; 58:121-45. [PMID: 25835096 DOI: 10.1016/j.plipres.2015.03.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 03/06/2015] [Accepted: 03/23/2015] [Indexed: 12/12/2022]
Abstract
Despite the growing interest in deciphering the causes and consequences of obesity-related disorders, the mechanisms linking fat intake to bone behaviour remain unclear. Since bone fractures are widely associated with increased morbidity and mortality, most notably in elderly and obese people, bone health has become a major social and economic issue. Consistently, public health system guidelines have encouraged low-fat diets in order to reduce associated complications. However, from a bone point of view, mechanisms linking fat intake to bone alteration remain quite controversial. Thus, after more than a decade of dedicated studies, this timely review offers a comprehensive overview of the relationships between bone and fatty acids. Using clinical evidences as a starting-point to more complex molecular elucidation, this work highlights the complexity of the system and reveals that bone alteration that cannot be solved simply by taking ω-3 pills. Fatty acid effects on bone metabolism can be both direct and indirect and require integrated investigations. Furthermore, even at the level of a single cell, one fatty acid is able to trigger several different independent pathways (receptors, metabolites…) which may all have a say in the final cellular metabolic response.
Collapse
Affiliation(s)
- Fabien Wauquier
- INRA, UMR 1019, UNH, CRNH Auvergne, F-63009 Clermont-Ferrand, France; Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, BP 10448, F-63000 Clermont-Ferrand, France; Equipe Alimentation, Squelette et Métabolismes, France
| | - Laurent Léotoing
- INRA, UMR 1019, UNH, CRNH Auvergne, F-63009 Clermont-Ferrand, France; Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, BP 10448, F-63000 Clermont-Ferrand, France; Equipe Alimentation, Squelette et Métabolismes, France
| | - Claire Philippe
- INRA, UMR 1019, UNH, CRNH Auvergne, F-63009 Clermont-Ferrand, France; Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, BP 10448, F-63000 Clermont-Ferrand, France; Equipe Alimentation, Squelette et Métabolismes, France
| | - Mélanie Spilmont
- INRA, UMR 1019, UNH, CRNH Auvergne, F-63009 Clermont-Ferrand, France; Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, BP 10448, F-63000 Clermont-Ferrand, France; Equipe Alimentation, Squelette et Métabolismes, France
| | - Véronique Coxam
- INRA, UMR 1019, UNH, CRNH Auvergne, F-63009 Clermont-Ferrand, France; Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, BP 10448, F-63000 Clermont-Ferrand, France; Equipe Alimentation, Squelette et Métabolismes, France
| | - Yohann Wittrant
- INRA, UMR 1019, UNH, CRNH Auvergne, F-63009 Clermont-Ferrand, France; Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, BP 10448, F-63000 Clermont-Ferrand, France; Equipe Alimentation, Squelette et Métabolismes, France.
| |
Collapse
|
146
|
Cai Y, Lydic TA, Turkette T, Reid GE, Olson LK. Impact of alogliptin and pioglitazone on lipid metabolism in islets of prediabetic and diabetic Zucker Diabetic Fatty rats. Biochem Pharmacol 2015; 95:46-57. [PMID: 25801003 DOI: 10.1016/j.bcp.2015.03.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 03/13/2015] [Indexed: 12/30/2022]
Abstract
Prolonged exposure of pancreatic beta (β) cells to elevated glucose and free fatty acids (FFA) as occurs in type 2 diabetes results in loss of β cell function and survival. In Zucker Diabetic Fatty (ZDF) rats, β cell failure is associated with increased triacylglyceride (TAG) synthesis and disruption of the glycerolipid/FFA (GL/FFA) cycle, a critical arm of glucose-stimulated insulin secretion (GSIS). The aim of this study was to determine the impact of activation of PPARγ and increased incretin action via dipeptidyl-peptidase inhibition using pioglitazone and/or alogliptin, respectively, on islet lipid metabolism in prediabetic and diabetic ZDF rats. Transition of control prediabetic ZDF rats to diabetes was associated with reduced plasma insulin levels, reduced islet insulin content and GSIS, reduced stearoyl-CoA desaturase 2 (SCD 2) expression, and increased islet TAG, diacylglyceride (DAG) and ceramides species containing saturated FA. Treatment of prediabetic ZDF rats with a combination of pioglitazone and alogliptin, but not individually, prevented the transition to diabetes and was associated with marked lowering of islet TAG and DAG levels. Pioglitazone and alogliptin, however, did not restore SCD2 expression, the degree of FA saturation in TAG, DAG or ceramides, islet insulin content, or lower ceramide levels. These findings are consistent with activation of PPARγ and increased incretin action working in concert to restore GL/FFA cycle in β cells of ZDF rats. Restoration of the GL/FFA cycle without correcting islet FA desaturation, production of islet ceramides, and/or insulin sensitivity, however, may place these islets at risk for β cell failure.
Collapse
Affiliation(s)
- Ying Cai
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA.
| | - Todd A Lydic
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, USA.
| | - Thomas Turkette
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA.
| | - Gavin E Reid
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, USA; Department of Chemistry, Michigan State University, East Lansing, MI 48824 USA.
| | - L Karl Olson
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
147
|
Oh YS. Mechanistic insights into pancreatic beta-cell mass regulation by glucose and free fatty acids. Anat Cell Biol 2015; 48:16-24. [PMID: 25806118 PMCID: PMC4371177 DOI: 10.5115/acb.2015.48.1.16] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Accepted: 02/04/2015] [Indexed: 01/14/2023] Open
Abstract
Pancreatic islets are responsible for blood glucose homeostasis. Reduced numbers of functional (insulin-secreting) beta-cells in pancreatic islets underlies diabetes. Restoration of the secretion of the proper amount of insulin is a goal. Beta-cell mass is increased by neogenesis, proliferation and cell hypertrophy, and is decreased by beta-cell death primarily through apoptosis. Many hormones and nutrients affect beta-cell mass, and glucose and free fatty acid are thought to be the most important determinants of beta-cell equilibrium. A number of molecular pathways have been implicated in beta-cell mass regulation and have been studied. This review will focus on the role of the principle metabolites, glucose and free fatty acid, and the downstream signaling pathways regulating beta-cell mass by these metabolites.
Collapse
Affiliation(s)
- Yoon Sin Oh
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea. ; Gachon Medical Research Institute, Gil Hospital, Incheon, Korea
| |
Collapse
|
148
|
Kim IS, Yang SY, Han JH, Jung SH, Park HS, Myung CS. Differential Gene Expression in GPR40-Overexpressing Pancreatic β-cells Treated with Linoleic Acid. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2015; 19:141-9. [PMID: 25729276 PMCID: PMC4342734 DOI: 10.4196/kjpp.2015.19.2.141] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Revised: 01/05/2015] [Accepted: 01/07/2015] [Indexed: 01/20/2023]
Abstract
"G protein-coupled receptor 40" (GPR40), a receptor for long-chain fatty acids, mediates the stimulation of glucose-induced insulin secretion. We examined the profiles of differential gene expression in GPR40-activated cells treated with linoleic acid, and finally predicted the integral pathways of the cellular mechanism of GPR40-mediated insulinotropic effects. After constructing a GPR40-overexpressing stable cell line (RIN-40) from the rat pancreatic β-cell line RIN-5f, we determined the gene expression profiles of RIN-5f and RIN-40. In total, 1004 genes, the expression of which was altered at least twofold, were selected in RIN-5f versus RIN-40. Moreover, the differential genetic profiles were investigated in RIN-40 cells treated with 30 µM linoleic acid, which resulted in selection of 93 genes in RIN-40 versus RIN-40 treated with linoleic acid. Based on the Kyoto Encyclopedia of Genes and Genomes Pathway (KEGG, http://www.genome.jp/kegg/), sets of genes induced differentially by treatment with linoleic acid in RIN-40 cells were found to be related to mitogen-activated protein (MAP) kinase- and neuroactive ligand-receptor interaction pathways. A gene ontology (GO) study revealed that more than 30% of the genes were associated with signal transduction and cell proliferation. Thus, this study elucidated a gene expression pattern relevant to the signal pathways that are regulated by GPR40 activation during the acute period. Together, these findings increase our mechanistic understanding of endogenous molecules associated with GPR40 function, and provide information useful for identification of a target for the management of type 2 diabetes mellitus.
Collapse
Affiliation(s)
- In-Su Kim
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon 305-764, Korea
| | - So-Young Yang
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon 305-764, Korea. ; Institute of Drug Research & Development, Chungnam National University, Daejeon 305-764, Korea
| | - Joo-Hui Han
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon 305-764, Korea
| | - Sang-Hyuk Jung
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon 305-764, Korea
| | - Hyun-Soo Park
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon 305-764, Korea
| | - Chang-Seon Myung
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon 305-764, Korea. ; Institute of Drug Research & Development, Chungnam National University, Daejeon 305-764, Korea
| |
Collapse
|
149
|
Wang X, Zhao T, Yang B, Li Z, Cui J, Dai Y, Qiu Q, Qiang H, Huang W, Qian H. Synthesis and biological evaluation of phenoxyacetic acid derivatives as novel free fatty acid receptor 1 agonists. Bioorg Med Chem 2015; 23:132-40. [DOI: 10.1016/j.bmc.2014.11.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 11/08/2014] [Accepted: 11/11/2014] [Indexed: 10/24/2022]
|
150
|
Alejandro EU, Gregg B, Blandino-Rosano M, Cras-Méneur C, Bernal-Mizrachi E. Natural history of β-cell adaptation and failure in type 2 diabetes. Mol Aspects Med 2014; 42:19-41. [PMID: 25542976 DOI: 10.1016/j.mam.2014.12.002] [Citation(s) in RCA: 163] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 11/04/2014] [Accepted: 12/05/2014] [Indexed: 02/07/2023]
Abstract
Type 2 diabetes mellitus (T2D) is a complex disease characterized by β-cell failure in the setting of insulin resistance. The current evidence suggests that genetic predisposition, and environmental factors can impair the capacity of the β-cells to respond to insulin resistance and ultimately lead to their failure. However, genetic studies have demonstrated that known variants account for less than 10% of the overall estimated T2D risk, suggesting that additional unidentified factors contribute to susceptibility of this disease. In this review, we will discuss the different stages that contribute to the development of β-cell failure in T2D. We divide the natural history of this process in three major stages: susceptibility, β-cell adaptation and β-cell failure, and provide an overview of the molecular mechanisms involved. Further research into mechanisms will reveal key modulators of β-cell failure and thus identify possible novel therapeutic targets and potential interventions to protect against β-cell failure.
Collapse
Affiliation(s)
- Emilyn U Alejandro
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, Brehm Center for Diabetes Research, University of Michigan, Ann Arbor, MI, USA
| | - Brigid Gregg
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Manuel Blandino-Rosano
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, Brehm Center for Diabetes Research, University of Michigan, Ann Arbor, MI, USA
| | - Corentin Cras-Méneur
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, Brehm Center for Diabetes Research, University of Michigan, Ann Arbor, MI, USA
| | - Ernesto Bernal-Mizrachi
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, Brehm Center for Diabetes Research, University of Michigan, Ann Arbor, MI, USA; VA Ann Arbor Healthcare System, Ann Arbor, MI, USA.
| |
Collapse
|