1
|
Soliman AR, Magd Eldin Saleem H, El Meligi AAH, Naguib M, Sobh Mohamed R, Abdelaziz GR, Rakha M, Abdelghaffar S, Hamed AE, Hammad HAERS, Mahmoud EO, Shaltout I. Metabolic/bariatric surgery optimization: a position statement by Arabic association for the study of diabetes and metabolism (AASD). Diabetol Metab Syndr 2025; 17:37. [PMID: 39881371 PMCID: PMC11776182 DOI: 10.1186/s13098-024-01564-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 12/20/2024] [Indexed: 01/31/2025] Open
Abstract
For patients considering bariatric surgery, it is essential to have clear answers to common questions to ensure the success of the procedure. Patients should understand that surgery is not a quick fix but a tool that must be complemented by lifestyle changes, including dietary adjustments and regular physical activity. The procedure carries potential risks that should be weighed against the potential benefits. Health authorities play a critical role in ensuring that bariatric surgery is performed under the highest standards of care. Recommendations are provided to determine who is an appropriate candidate for surgery, what preoperative evaluations are necessary, and how to monitor patients postoperatively to maximize outcomes and minimize risks. Additionally, authorities are responsible for ensuring access to follow-up care, including nutritional support and psychological counseling, which are vital for the long-term success of bariatric surgery.Understanding these aspects by both patients and decision-makers is critical before proceeding with bariatric surgery. The following questions guide patients and healthcare professionals in making informed decisions about the procedure and managing the expectations and outcomes associated with bariatric surgery.
Collapse
Affiliation(s)
| | - Hesham Magd Eldin Saleem
- Faculty of Medicine,Cairo University,Internal Medicine Diabetes and Endocrinology Department, Cairo, Egypt
| | - Amr Abel Hady El Meligi
- Faculty of Medicine,Cairo University,Internal Medicine Diabetes and Endocrinology Department, Cairo, Egypt
| | - Mervat Naguib
- Faculty of Medicine,Cairo University,Internal Medicine Diabetes and Endocrinology Department, Cairo, Egypt
| | - Rasha Sobh Mohamed
- Faculty of Medicine,Cairo University,Internal Medicine Diabetes and Endocrinology Department, Cairo, Egypt
| | - Ghada Rabie Abdelaziz
- Faculty of Medicine,Cairo University,Internal Medicine Diabetes and Endocrinology Department, Cairo, Egypt
| | - Maha Rakha
- Faculty of Medicine,Cairo University,Internal Medicine Diabetes and Endocrinology Department, Cairo, Egypt
| | - Shereen Abdelghaffar
- Faculty of Medicine,Cairo University, Pediatric Diabetes and Endocrinology Department, Cairo, Egypt
| | | | | | - Eman O Mahmoud
- Faculty of Medicine,Cairo University,Internal Medicine Diabetes and Endocrinology Department, Cairo, Egypt.
| | - Inass Shaltout
- Faculty of Medicine,Cairo University,Internal Medicine Diabetes and Endocrinology Department, Cairo, Egypt
| |
Collapse
|
2
|
Coomans de Brachène A, Alvelos MI, Szymczak F, Zimath PL, Castela A, Marmontel de Souza B, Roca Rivada A, Marín-Cañas S, Yi X, Op de Beeck A, Morgan NG, Sonntag S, Jawurek S, Title AC, Yesildag B, Pattou F, Kerr-Conte J, Montanya E, Nacher M, Marselli L, Marchetti P, Richardson SJ, Eizirik DL. Interferons are key cytokines acting on pancreatic islets in type 1 diabetes. Diabetologia 2024; 67:908-927. [PMID: 38409439 DOI: 10.1007/s00125-024-06106-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 01/03/2024] [Indexed: 02/28/2024]
Abstract
AIMS/HYPOTHESIS The proinflammatory cytokines IFN-α, IFN-γ, IL-1β and TNF-α may contribute to innate and adaptive immune responses during insulitis in type 1 diabetes and therefore represent attractive therapeutic targets to protect beta cells. However, the specific role of each of these cytokines individually on pancreatic beta cells remains unknown. METHODS We used deep RNA-seq analysis, followed by extensive confirmation experiments based on reverse transcription-quantitative PCR (RT-qPCR), western blot, histology and use of siRNAs, to characterise the response of human pancreatic beta cells to each cytokine individually and compared the signatures obtained with those present in islets of individuals affected by type 1 diabetes. RESULTS IFN-α and IFN-γ had a greater impact on the beta cell transcriptome when compared with IL-1β and TNF-α. The IFN-induced gene signatures have a strong correlation with those observed in beta cells from individuals with type 1 diabetes, and the level of expression of specific IFN-stimulated genes is positively correlated with proteins present in islets of these individuals, regulating beta cell responses to 'danger signals' such as viral infections. Zinc finger NFX1-type containing 1 (ZNFX1), a double-stranded RNA sensor, was identified as highly induced by IFNs and shown to play a key role in the antiviral response in beta cells. CONCLUSIONS/INTERPRETATION These data suggest that IFN-α and IFN-γ are key cytokines at the islet level in human type 1 diabetes, contributing to the triggering and amplification of autoimmunity.
Collapse
Affiliation(s)
| | - Maria Ines Alvelos
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Florian Szymczak
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Priscila L Zimath
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Angela Castela
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | | | - Arturo Roca Rivada
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Sandra Marín-Cañas
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Xiaoyan Yi
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Anne Op de Beeck
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Noel G Morgan
- Islet Biology Exeter (IBEx), Exeter Centre of Excellence for Diabetes Research (EXCEED), Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter, UK
| | - Sebastian Sonntag
- InSphero AG, Schlieren, Switzerland
- University of Applied Sciences and Arts Northwestern Switzerland, Basel, Switzerland
| | | | | | | | - François Pattou
- European Genomic Institute for Diabetes, UMR 1190 Translational Research for Diabetes, Inserm, CHU Lille, University of Lille, Lille, France
| | - Julie Kerr-Conte
- European Genomic Institute for Diabetes, UMR 1190 Translational Research for Diabetes, Inserm, CHU Lille, University of Lille, Lille, France
| | - Eduard Montanya
- Hospital Universitari Bellvitge, Bellvitge Biomedical Research Institute (IDIBELL), Centro de Investigación Biomédica en Red Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) and University of Barcelona, Barcelona, Spain
| | - Montserrat Nacher
- Hospital Universitari Bellvitge, Bellvitge Biomedical Research Institute (IDIBELL), Centro de Investigación Biomédica en Red Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) and University of Barcelona, Barcelona, Spain
| | - Lorella Marselli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Sarah J Richardson
- Islet Biology Exeter (IBEx), Exeter Centre of Excellence for Diabetes Research (EXCEED), Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter, UK
| | - Decio L Eizirik
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium.
| |
Collapse
|
3
|
Enkaku A, Chujo D, Kamigishi M, Inagawa S, Matsukoshi S, Sakai W, Takikawa A, Fujisaka S, Tobe K. Short-term recovery of insulin secretion in response to a meal is associated with future glycemic control in type 2 diabetes patients. J Diabetes Investig 2024; 15:437-448. [PMID: 38151917 PMCID: PMC10981139 DOI: 10.1111/jdi.14129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 11/12/2023] [Accepted: 11/30/2023] [Indexed: 12/29/2023] Open
Abstract
AIMS/INTRODUCTION Endogenous insulin secretion could be recovered by improving hyperglycemia in patients with type 2 diabetes. This study aimed to investigate the association between short-term recovery of insulin secretion during hospitalization and clinical background or future glycemic control in patients with type 2 diabetes. MATERIALS AND METHODS A total of 127 patients with type 2 diabetes were included. The recovery of endogenous insulin secretion was determined using the following indices: index A: fasting C-peptide index (CPI) at discharge - fasting CPI on admission; index B: postprandial CPI at discharge - postprandial CPI on admission; and index C: Δ C-peptide immunoreactivity (CPR) (postprandial CPR - fasting CPR) at discharge - ΔCPR on admission. We examined the associations of each index with clinical background and future glycemic control measured by glycosylated hemoglobin and continuous glucose monitoring. RESULTS Using index A and B, the age was significantly younger, whereas BMI and visceral fat area were significantly higher in the high-recovery group than in the low-recovery group. Changes in glycosylated hemoglobin levels were significantly greater at 6 and 12 months in the high-recovery group in the analysis of index C. The receiver operating characteristic curve analysis identified the index B and index C as indicators to predict glycosylated hemoglobin <7.0% at 6 months after discharge. Furthermore, index C was positively correlated with the time in the target glucose range, and inversely correlated with the standard deviation of glucose at 3 and 12 months after discharge. CONCLUSIONS Short-term recovery of insulin secretion in response to a meal during hospitalization, evaluated with the index-C, might predict future glycemic control.
Collapse
Affiliation(s)
- Asako Enkaku
- Department of Diabetes, Metabolism, and EndocrinologyToyama University HospitalToyamaJapan
| | - Daisuke Chujo
- Department of Diabetes, Metabolism, and EndocrinologyToyama University HospitalToyamaJapan
- Center for Clinical ResearchToyama University HospitalToyamaJapan
| | - Miki Kamigishi
- Department of Diabetes, Metabolism, and EndocrinologyToyama University HospitalToyamaJapan
| | - Shinya Inagawa
- Department of Diabetes, Metabolism, and EndocrinologyToyama University HospitalToyamaJapan
| | - Shinnosuke Matsukoshi
- Department of Diabetes, Metabolism, and EndocrinologyToyama University HospitalToyamaJapan
| | - Waka Sakai
- Department of Diabetes, Metabolism, and EndocrinologyToyama University HospitalToyamaJapan
| | - Akiko Takikawa
- Department of Diabetes, Metabolism, and EndocrinologyToyama University HospitalToyamaJapan
| | - Shiho Fujisaka
- Department of Diabetes, Metabolism, and EndocrinologyToyama University HospitalToyamaJapan
| | - Kazuyuki Tobe
- Department of Diabetes, Metabolism, and EndocrinologyToyama University HospitalToyamaJapan
| |
Collapse
|
4
|
Firdos, Pramanik T, Verma P, Mittal A. (Re-)Viewing Role of Intracellular Glucose Beyond Extracellular Regulation of Glucose-Stimulated Insulin Secretion by Pancreatic Cells. ACS OMEGA 2024; 9:11755-11768. [PMID: 38496986 PMCID: PMC10938456 DOI: 10.1021/acsomega.3c09171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/31/2024] [Accepted: 02/07/2024] [Indexed: 03/19/2024]
Abstract
For glucose-stimulated insulin secretion (GSIS) by pancreatic β-cells in animals, it is believed that ATP generated from glucose metabolism is primarily responsible. However, this ignores two well-established aspects in literature: (a) intracellular ATP generation from other sources resulting in an overall pool of ATP, regardless of the original source, and (b) that intracellular glucose transport is 10- to 100-fold higher than intracellular glucose phosphorylation in β-cells. The latter especially provides an earlier unaddressed, but highly appealing, observation pertaining to (at least transient) the presence of intracellular glucose molecules. Could these intracellular glucose molecules be responsible for the specificity of GSIS to glucose (instead of the widely believed ATP production from its metabolism)? In this work, we provide a comprehensive compilation of literature on glucose and GSIS using various cellular systems - all studies focus only on the extracellular role of glucose in GSIS. Further, we carried out a comprehensive analysis of differential gene expression in Mouse Insulinoma 6 (MIN6) cells, exposed to low and high extracellular glucose concentrations (EGC), from the existing whole transcriptome data. The expression of other genes involved in glycolysis, Krebs cycle, and electron transport chain was found to be unaffected by EGC, except Gapdh, Atp6v0a4, and Cox20. Remarkably, 3 upregulated genes (Atp6v0a4, Cacnb4, Kif11) in high EGC were identified to have an association with cellular secretion. Using glucose as a possible ligand for the 3 proteins, computational investigations were carried out (that will require future 'wet validation', both in vitro and in vivo, e.g., using primary islets and animal models). The glucose-affinity/binding scores (in kcal/mol) obtained were also compared with glucose binding scores for positive controls (GCK and GLUT2), along with negative controls (RPA1, KU70-80, POLA1, ACAA1A, POLR1A). The binding affinity scores of glucose molecules for the 3 proteins were found to be closer to positive controls. Therefore, we report the glucose binding ability of 3 secretion-related proteins and a possible direct role of intracellular glucose molecules in GSIS.
Collapse
Affiliation(s)
- Firdos
- Kusuma
School of Biological Sciences, Indian Institute
of Technology Delhi (IIT Delhi), Hauz Khas, New Delhi 110016, India
| | - Tapabrata Pramanik
- Kusuma
School of Biological Sciences, Indian Institute
of Technology Delhi (IIT Delhi), Hauz Khas, New Delhi 110016, India
| | - Prachi Verma
- Kusuma
School of Biological Sciences, Indian Institute
of Technology Delhi (IIT Delhi), Hauz Khas, New Delhi 110016, India
| | - Aditya Mittal
- Kusuma
School of Biological Sciences, Indian Institute
of Technology Delhi (IIT Delhi), Hauz Khas, New Delhi 110016, India
- Supercomputing
Facility for Bioinformatics and Computational Biology (SCFBio), IIT Delhi, Hauz Khas, New Delhi, 110016, India
| |
Collapse
|
5
|
Liu J, Fu H, Kang F, Ning G, Ni Q, Wang W, Wang Q. β-Cell glucokinase expression was increased in type 2 diabetes subjects with better glycemic control. J Diabetes 2023; 15:409-418. [PMID: 36942376 PMCID: PMC10172022 DOI: 10.1111/1753-0407.13380] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 02/22/2023] [Accepted: 02/26/2023] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND Type 2 diabetes (T2D) is characterized by a progressive deterioration of β-cell function with a continuous decline in insulin secretion. Glucokinase (GCK) facilitates the rate-limiting step of glycolysis in pancreatic β-cells, to acquire the proper glucose-stimulated insulin secretion. Multiple glucokinase activators (GKAs) have been developed and clinically tested. However, the dynamic change of human pancreatic GCK expression during T2D progression has not been investigated. METHODS We evaluated GCK expression by measuring the average immunoreactivity of GCK in insulin+ or glucagon+ cells from pancreatic sections of 11 nondiabetic subjects (ND), 10 subjects with impaired fasting glucose (IFG), 9 with well-controlled T2D (wT2D), and 5 individuals with poorly controlled T2D (uT2D). We also assessed the relationship between GCK expression and adaptive unfolded protein response (UPR) in human diabetic β-cells. RESULTS We did not detect changes of GCK expression in IFG islets. However, we found β-cell GCK levels were significantly increased in T2D with adequate glucose control (wT2D) but not in T2D with poor glucose control (uT2D). Furthermore, there was a strong positive correlation between GCK expression and adaptive UPR (spliced X-box binding protein 1 [XBP1s] and activating transcription factor 4 [ATF4]), as well as functional maturity marker (urocortin-3 [UCN3]) in human diabetic β-cells. CONCLUSIONS Our study demonstrates that inductions of GCK enhanced adaptive UPR and UCN3 in human β-cells, which might be an adaptive mechanism during T2D progression. This finding provides a rationale for exploring novel molecules that activate β-cell GCK and thereby improve pharmacological treatment of T2D.
Collapse
Affiliation(s)
- Jingwen Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Hui Fu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Fuyun Kang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Sino‐French Research Center for Life Sciences and Genomics Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Qicheng Ni
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Qidi Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Sino‐French Research Center for Life Sciences and Genomics Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
6
|
Giusti L, Tesi M, Ciregia F, Marselli L, Zallocco L, Suleiman M, De Luca C, Del Guerra S, Zuccarini M, Trerotola M, Eizirik DL, Cnop M, Mazzoni MR, Marchetti P, Lucacchini A, Ronci M. The Protective Action of Metformin against Pro-Inflammatory Cytokine-Induced Human Islet Cell Damage and the Mechanisms Involved. Cells 2022; 11:2465. [PMID: 35954309 PMCID: PMC9368307 DOI: 10.3390/cells11152465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/04/2022] [Accepted: 08/06/2022] [Indexed: 11/24/2022] Open
Abstract
Metformin, a drug widely used in type 2 diabetes (T2D), has been shown to protect human β-cells exposed to gluco- and/or lipotoxic conditions and those in islets from T2D donors. We assessed whether metformin could relieve the human β-cell stress induced by pro-inflammatory cytokines (which mediate β-cells damage in type 1 diabetes, T1D) and investigated the underlying mechanisms using shotgun proteomics. Human islets were exposed to 50 U/mL interleukin-1β plus 1000 U/mL interferon-γ for 48 h, with or without 2.4 µg/mL metformin. Glucose-stimulated insulin secretion (GSIS) and caspase 3/7 activity were studied, and a shotgun label free proteomics analysis was performed. Metformin prevented the reduction of GSIS and the activation of caspase 3/7 induced by cytokines. Proteomics analysis identified more than 3000 proteins in human islets. Cytokines alone altered the expression of 244 proteins (145 up- and 99 down-regulated), while, in the presence of metformin, cytokine-exposure modified the expression of 231 proteins (128 up- and 103 downregulated). Among the proteins inversely regulated in the two conditions, we found proteins involved in vesicle motility, defense against oxidative stress (including peroxiredoxins), metabolism, protein synthesis, glycolysis and its regulation, and cytoskeletal proteins. Metformin inhibited pathways linked to inflammation, immune reactions, mammalian target of rapamycin (mTOR) signaling, and cell senescence. Some of the changes were confirmed by Western blot. Therefore, metformin prevented part of the deleterious actions of pro-inflammatory cytokines in human β-cells, which was accompanied by islet proteome modifications. This suggests that metformin, besides use in T2D, might be considered for β-cell protection in other types of diabetes, possibly including early T1D.
Collapse
Affiliation(s)
- Laura Giusti
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy
| | - Marta Tesi
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Federica Ciregia
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, University of Liège, 4000 Liège, Belgium
| | - Lorella Marselli
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | | | - Mara Suleiman
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Carmela De Luca
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Silvia Del Guerra
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Mariachiara Zuccarini
- Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, 66100 Chieti, Italy
- Department of Medical, Oral and Biotechnological Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Marco Trerotola
- Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, 66100 Chieti, Italy
- Department of Medical, Oral and Biotechnological Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Decio L. Eizirik
- ULB Center for Diabetes Research, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Miriam Cnop
- ULB Center for Diabetes Research, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | | | - Piero Marchetti
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Antonio Lucacchini
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Maurizio Ronci
- Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, 66100 Chieti, Italy
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| |
Collapse
|
7
|
Ahrén B. The Glucose Sensitivity of Insulin Secretion-Lessons from In Vivo and In Vitro Studies in Mice. Biomolecules 2022; 12:biom12070976. [PMID: 35883532 PMCID: PMC9312818 DOI: 10.3390/biom12070976] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 06/27/2022] [Accepted: 07/08/2022] [Indexed: 02/05/2023] Open
Abstract
This study explored the relationship between the glucose dose and insulin response from beta cells in vivo and in vitro in mice. Glucose was administered intravenously at different dose levels (from 0 to 0.75 g/kg) in anesthetized C57BL/6J mice, and the glucose and insulin concentrations were determined in samples taken after 50 min. Furthermore, freshly isolated mouse islets were incubated for 60 min in the presence of different concentrations of glucose (from 2.8 to 22.2 mmol/L) and insulin levels were analyzed in the medium. It was found that insulin levels increased after an intravenous injection of glucose with the maximal increase seen after 0.35 g/kg with no further increase after 0.5 or 0.75 g/kg. The acute increase in insulin levels (during the first 5 min) and the maximum glucose level (achieved after 1 min) showed a curvilinear relation with the half-maximal increase in insulin levels achieved at 11.4 mmol/L glucose and the maximal increase in insulin levels at 22.0 mmol/L glucose. In vitro, there was also a curvilinear relation between glucose concentrations and insulin secretion. Half maximal increase in insulin concentrations was achieved at 12.5 mmol/L glucose and the maximal increase in insulin concentrations was achieved at 21.5 mmol/L. Based on these data, we concluded that the glucose-insulin relation was curvilinear both in vivo and in vitro in mice with similar characteristics in relation to which glucose levels that achieve half-maximal and maximal increases in insulin secretion. Besides the new knowledge of knowing these relations, the results have consequences on how to design studies on insulin secretion to obtain the most information.
Collapse
Affiliation(s)
- Bo Ahrén
- Department of Clinical Sciences Lund, Lund University, 22185 Lund, Sweden
| |
Collapse
|
8
|
Boontem P, Yamashima T. Hydroxynonenal causes Langerhans cell degeneration in the pancreas of Japanese macaque monkeys. PLoS One 2021; 16:e0245702. [PMID: 34748564 PMCID: PMC8575276 DOI: 10.1371/journal.pone.0245702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 09/18/2021] [Indexed: 11/21/2022] Open
Abstract
Background For their functions of insulin biosynthesis and glucose- and fatty acid- mediated insulin secretion, Langerhans β-cells require an intracellular milieu rich in oxygen. This requirement makes β-cells, with their constitutively low antioxidative defense, susceptible to the oxidative stress. Although much progress has been made in identifying its molecular basis in experimental systems, whether the oxidative stress due to excessive fatty acids plays a crucial role in the Langerhans cell degeneration in primates is still debated. Methods Focusing on Hsp70.1, which has dual functions as molecular chaperone and lysosomal stabilizer, the mechanism of lipotoxicity to Langerhans cells was studied using macaque monkeys after the consecutive injections of the lipid peroxidation product ‘hydroxynonenal’. Based on the ‘calpain-cathepsin hypothesis’ formulated in 1998, calpain activation, Hsp70.1 cleavage, and lysosomal integrity were studied by immunofluorescence histochemistry, electron microscopy, and Western blotting. Results Light microscopy showed more abundant vacuole formation in the hydroxynonenal-treated islet cells than the control cells. Electron microscopy showed that vacuolar changes, which were identified as enlarged rough ER, occurred mainly in β-cells followed by δ-cells. Intriguingly, both cell types showed a marked decrease in insulin and somatostatin granules. Furthermore, they exhibited marked increases in peroxisomes, autophagosomes/autolysosomes, lysosomal and peroxisomal membrane rupture/permeabilization, and mitochondrial degeneration. Disrupted peroxisomes were often localized in the close vicinity of degenerating mitochondria or autolysosomes. Immunofluorescence histochemical analysis showed an increased co-localization of activated μ-calpain and Hsp70.1 with the extralysosomal release of cathepsin B. Western blotting showed increases in μ-calpain activation, Hsp70.1 cleavage, and expression of the hydroxynonenal receptor GPR109A. Conclusions Taken together, these data implicate hydroxynonenal in both oxidation of Hsp70.1 and activation of μ-calpain. The calpain-mediated cleavage of the carbonylated Hsp70.1, may cause lysosomal membrane rupture/permeabilization. The low defense of primate Langerhans cells against hydroxynonenal and peroxisomally-generated hydrogen peroxide, was presumably overwhelmed to facilitate cell degeneration.
Collapse
Affiliation(s)
| | - Tetsumori Yamashima
- Departments of Cell Metabolism and Nutrition, Kanazawa, Japan
- Psychiatry and Behavioral Science, Kanazawa, Japan
- * E-mail:
| |
Collapse
|
9
|
Chu KY, Mellet N, Thai LM, Meikle PJ, Biden TJ. Short-term inhibition of autophagy benefits pancreatic β-cells by augmenting ether lipids and peroxisomal function, and by countering depletion of n-3 polyunsaturated fatty acids after fat-feeding. Mol Metab 2020; 40:101023. [PMID: 32504884 PMCID: PMC7322075 DOI: 10.1016/j.molmet.2020.101023] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 04/29/2020] [Accepted: 05/14/2020] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE Investigations of autophagy in β-cells have usually focused on its homeostatic function. More dynamic roles in inhibiting glucose-stimulated insulin secretion (GSIS), potentially involving remodelling of cellular lipids, have been suggested from in vitro studies but not evaluated in vivo. METHODS We employed temporally-regulated deletion of the essential autophagy gene, Atg7, in β-cells. Mice were fed chow or high-fat diets (HFD), in conjunction with deletion of Atg7 for the last 3 weeks (short-term model) or 9 weeks (long-term model). Standard in vivo metabolic phenotyping was undertaken, and 450 lipid species in islets quantified ex vivo using mass spectroscopy (MS). MIN6 cells were also employed for lipidomics and secretory interventions. RESULTS β-cell function was impaired by inhibiting autophagy in the longer-term, but conversely improved by 3-week deletion of Atg7, specifically under HFD conditions. This was accompanied by augmented GSIS ex vivo. Surprisingly, the HFD had minimal effect on sphingolipid and neutral lipid species, but modulated >100 phospholipids and ether lipids, and markedly shifted the profile of polyunsaturated fatty acid (PUFA) sidechains from n3 to n6 forms. These changes were partially countered by Atg7 deletion, consistent with an accompanying upregulation of the PUFA elongase enzyme, Elovl5. Loss of Atg7 separately augmented plasmalogens and alkyl lipids, in association with increased expression of Lonp2, a peroxisomal chaperone/protease that facilitates maturation of ether lipid synthetic enzymes. Depletion of PUFAs and ether lipids was also observed in MIN6 cells chronically exposed to oleate (more so than palmitate). GSIS was inhibited by knocking down Dhrs7b, which encodes an enzyme of peroxisomal ether lipid synthesis. Conversely, impaired GSIS due to oleate pre-treatment was selectively reverted by Dhrs7b overexpression. CONCLUSIONS A detrimental increase in n6:n3 PUFA ratios in ether lipids and phospholipids is revealed as a major response of β-cells to high-fat feeding. This is partially reversed by short-term inhibition of autophagy, which results in compensatory changes in peroxisomal lipid metabolism. The short-term phenotype is linked to improved GSIS, in contrast to the impairment seen with the longer-term inhibition of autophagy. The balance between these positive and negative inputs could help determine whether β-cells adapt or fail in response to obesity.
Collapse
Affiliation(s)
- Kwan Yi Chu
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW, 2010, Australia
| | - Natalie Mellet
- Baker Heart and Diabetes Institute, PO Box 6492, Melbourne, Vic, 3004, Australia
| | - Le May Thai
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW, 2010, Australia
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, PO Box 6492, Melbourne, Vic, 3004, Australia.
| | - Trevor J Biden
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW, 2010, Australia; St Vincent's Clinical School, Faculty of Medicine, The University of New South Wales, Sydney, NSW, Australia.
| |
Collapse
|
10
|
Iwata M, Kamura Y, Honoki H, Kobayashi K, Ishiki M, Yagi K, Fukushima Y, Takano A, Kato H, Murakami S, Higuchi K, Kobashi C, Fukuda K, Koshimizu Y, Tobe K. Family history of diabetes in both parents is strongly associated with impaired residual β-cell function in Japanese type 2 diabetes patients. J Diabetes Investig 2020; 11:564-572. [PMID: 31705736 PMCID: PMC7232274 DOI: 10.1111/jdi.13176] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 11/05/2019] [Accepted: 11/06/2019] [Indexed: 12/14/2022] Open
Abstract
AIMS/INTRODUCTION The objective of the present study was to clarify the association of the type and number of first-degree family history of diabetes (FHD) with the clinical characteristics, especially with residual β-cell function, in type 2 diabetes patients. MATERIALS AND METHODS A total of 1,131 type 2 diabetes patients were recruited and divided into four groups according to FHD information as follows: (i) patients without FHD (FHD-); (ii) those with at least one sibling who had diabetes without parental diabetes (FHD+); (iii) those with one parent (FHD++); or (iv) those with both parents (FHD+++) who had diabetes with or without a sibling with diabetes. RESULTS The percentages of the FHD-, FHD+, FHD++ and FHD+++ groups were 49.4%, 13.4%, 34.0% and 3.2%, respectively. Patients in the FHD++ and FHD+++ groups were significantly younger at the time of diabetes diagnosis (P < 0.001) than those in the FHD- and FHD+ groups, even after adjusting for confounding factors. In addition, the levels of insulin secretion were significantly lower in the patients in the FHD+, FHD++ and FHD+++ groups than those in the FHD- group (P < 0.05) after adjusting for confounding factors, and the patients in the FHD+++ group presented with the lowest levels of insulin secretion among the four groups. CONCLUSIONS Our results showed that in type 2 diabetes patients, the degree of the associations between FHD and clinical characteristics differs according to the number and the type of FHD. In particular, FHD in both parents is most strongly associated with impaired residual β-cell function.
Collapse
Affiliation(s)
- Minoru Iwata
- First Department of Internal MedicineFaculty of MedicineUniversity of ToyamaToyamaJapan
- Itoigawa Community Medical UnitToyama University HospitalToyamaJapan
| | - Yutaka Kamura
- First Department of Internal MedicineFaculty of MedicineUniversity of ToyamaToyamaJapan
| | - Hisae Honoki
- First Department of Internal MedicineFaculty of MedicineUniversity of ToyamaToyamaJapan
| | - Kaori Kobayashi
- First Department of Internal MedicineFaculty of MedicineUniversity of ToyamaToyamaJapan
| | - Manabu Ishiki
- First Department of Internal MedicineFaculty of MedicineUniversity of ToyamaToyamaJapan
- Center for Medical Education and Career DevelopmentUniversity of ToyamaToyamaJapan
| | - Kunimasa Yagi
- First Department of Internal MedicineFaculty of MedicineUniversity of ToyamaToyamaJapan
| | - Yasuo Fukushima
- Department of Internal MedicineAsahi General HospitalAsahi‐machiJapan
| | - Atsuko Takano
- Division of Endocrinology and MetabolismDepartment of Internal MedicineSaiseikai Takaoka HospitalTakaokaJapan
| | - Hiromi Kato
- Department of Internal MedicineJapan Community Health care Organization Takaoka Fushiki HospitalTakaokaJapan
| | - Shihou Murakami
- Division of Endocrinology and MetabolismDepartment of Internal MedicineToyama Rosai HospitalUozuJapan
| | - Kiyohiro Higuchi
- Department of Internal MedicineJA Niigata Kouseiren Itoigawa General HospitalItoigawaJapan
| | - Chikaaki Kobashi
- Department of Internal MedicineKamiichi General HospitalKamiichi‐machiJapan
| | | | | | - Kazuyuki Tobe
- First Department of Internal MedicineFaculty of MedicineUniversity of ToyamaToyamaJapan
| |
Collapse
|
11
|
Fang Z, Weng C, Li H, Tao R, Mai W, Liu X, Lu L, Lai S, Duan Q, Alvarez C, Arvan P, Wynshaw-Boris A, Li Y, Pei Y, Jin F, Li Y. Single-Cell Heterogeneity Analysis and CRISPR Screen Identify Key β-Cell-Specific Disease Genes. Cell Rep 2019; 26:3132-3144.e7. [PMID: 30865899 PMCID: PMC6573026 DOI: 10.1016/j.celrep.2019.02.043] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 05/03/2018] [Accepted: 02/12/2019] [Indexed: 01/13/2023] Open
Abstract
Identification of human disease signature genes typically requires samples from many donors to achieve statistical significance. Here, we show that single-cell heterogeneity analysis may overcome this hurdle by significantly improving the test sensitivity. We analyzed the transcriptome of 39,905 single islets cells from 9 donors and observed distinct β cell heterogeneity trajectories associated with obesity or type 2 diabetes (T2D). We therefore developed RePACT, a sensitive single-cell analysis algorithm to identify both common and specific signature genes for obesity and T2D. We mapped both β-cell-specific genes and disease signature genes to the insulin regulatory network identified from a genome-wide CRISPR screen. Our integrative analysis discovered the previously unrecognized roles of the cohesin loading complex and the NuA4/Tip60 histone acetyltransferase complex in regulating insulin transcription and release. Our study demonstrated the power of combining single-cell heterogeneity analysis and functional genomics to dissect the etiology of complex diseases.
Collapse
Affiliation(s)
- Zhou Fang
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Chen Weng
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Haiyan Li
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Ran Tao
- Center for Cancer and Immunology Research, Brain Tumor Institute, Children's National Medical Center, Washington, D.C. 20010, USA
| | - Weihua Mai
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; Department of Neurology, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Xiaoxiao Liu
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Leina Lu
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Sisi Lai
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Qing Duan
- Department of Biostatistics, Department of Genetics, Department of Computer Science, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Carlos Alvarez
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; The Biomedical Sciences Training Program (BSTP), School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Peter Arvan
- Division of Metabolism, Endocrinology, and Diabetes, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Anthony Wynshaw-Boris
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Yun Li
- Department of Biostatistics, Department of Genetics, Department of Computer Science, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Yanxin Pei
- Center for Cancer and Immunology Research, Brain Tumor Institute, Children's National Medical Center, Washington, D.C. 20010, USA
| | - Fulai Jin
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; Department of Population and Quantitative Health Sciences, Department of Electrical Engineering and Computer Science, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA.
| | - Yan Li
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA.
| |
Collapse
|
12
|
Lee PC, Tan HC, Pasupathy S, Ganguly S, Eng AKH, Nadkarni N, Tham KW. Effectiveness of bariatric surgery in diabetes prevention in high-risk Asian individuals. Singapore Med J 2018; 59:472-475. [PMID: 30310918 PMCID: PMC6158137 DOI: 10.11622/smedj.2018110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
INTRODUCTION Obesity is a key risk factor in the development of Type 2 diabetes mellitus (T2DM). Bariatric surgery causes a large amount of durable weight loss in those with clinically severe obesity. We reported the effect of weight loss via bariatric surgery on DM prevention in those at high risk of developing DM. METHODS This was a retrospective cohort study of 44 patients with obesity (mean body mass index 43.8 kg/m2) and pre-DM who underwent bariatric surgery and were followed up for up to three years. We also reviewed a non-surgical cohort of patients with obesity and pre-DM seen at the weight management clinic. RESULTS 91% of patients attained normal glycaemic status at one year after bariatric surgery. At the three-year follow-up, 87.5% of the patients maintained normoglycaemia. None of the patients developed T2DM after surgery. 26.9% of patients achieved absolute weight loss at one year after bariatric surgery and maintained this at two and three years post surgery (p < 0.001 vs. baseline). The homeostatic model assessment-insulin resistance index in patients also decreased from 5.50 at baseline to 1.20, 1.14 and 1.44 at one, two and three years, respectively (p < 0.001). CONCLUSION Bariatric surgery produces significant weight loss, and leads to reversion from the pre-diabetic state to normal glycaemic status and reduction of the incident DM rate in those with pre-DM and morbid obesity.
Collapse
Affiliation(s)
- Phong Ching Lee
- Department of Endocrinology, Singapore General Hospital, Singapore
| | - Hong Chang Tan
- Department of Endocrinology, Singapore General Hospital, Singapore
| | | | - Sonali Ganguly
- Department of Endocrinology, Singapore General Hospital, Singapore
| | - Alvin Kim Hock Eng
- Department of Upper Gastrointestinal and Bariatric Surgery, Singapore General Hospital, Singapore
| | - Nivedita Nadkarni
- Centre for Quantitative Medicine, Office of Clinical Sciences, Duke-NUS Medical School, Singapore
| | - Kwang Wei Tham
- Department of Endocrinology, Singapore General Hospital, Singapore
| |
Collapse
|
13
|
Kintoko K, Xu X, Lin X, Jiao Y, Wen Q, Chen Z, Wei J, Liang T, Huang R. Hypoglycaemic activity of 2-dodecyl-6-methoxycyclohexa-2,5-diene-1,4-dione in streptozotocin-induced diabetic mice through ameliorating metabolic function and regulating peroxisome proliferator-activated receptor γ. Arch Med Sci 2018; 14:1163-1172. [PMID: 30154901 PMCID: PMC6111351 DOI: 10.5114/aoms.2016.63285] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 08/12/2016] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Diabetes mellitus is characterized by hyperglycaemia causing changes in plasma lipoproteins, which leads to insulin resistance, secretion defects or both. The present study aimed to evaluate the ability of 2-dodecyl-6-methoxy-cyclohexa-2,5-diene-1,4-dione (DMDD) isolated from Averrhoa carambola L. roots to lower hyperglycaemia and to investigate its potential mechanism in diabetic mice. MATERIAL AND METHODS DMDD was isolated using a column chromatographic technique. Experimental mice were fed with a high-fat diet for a month and were intravenously injected with streptozotocin (80 mg/kg, single dose). Diabetic mice were orally administered DMDD (12.5, 25, 50 mg/kg) and 50 mg/kg pioglitazone for 15 days. Fasting blood glucose (FBG), fasting blood insulin (FINS), pancreatic insulin content, interleukin-6 (IL-6), tumour necrosis factor-α (TNF-α), as well as serum total cholesterol (TC), triglyceride (TG) and free fatty acid (FFA) were determined. Adipose tissue was assessed by histological examination, immunohistochemistry, western blot and reverse transcription-polymerase chain reaction methods. RESULTS DMDD significantly increased the insulin level (all p < 0.05). In contrast, FBG, IL-6, TNF-α, TC, TG and FFA were significantly decreased (all p < 0.05). However, DMDD induced the activation of adipocyte peroxisome proliferator-activated receptor γ (PPAR-γ), confirmed by increased protein and mRNA expression of PPAR-γ. CONCLUSIONS DMDD possessed hypoglycaemic activity due to its potential mechanism involving PPARγ-mediated adipocyte endocrine regulation.
Collapse
Affiliation(s)
- Kintoko Kintoko
- Pharmaceutical College, Guangxi Medical University, Guangxi, China
| | - Xiaohui Xu
- Pharmaceutical College, Guangxi Medical University, Guangxi, China
| | - Xing Lin
- Pharmaceutical College, Guangxi Medical University, Guangxi, China
| | - Yang Jiao
- Pharmaceutical College, Guangxi Medical University, Guangxi, China
| | - Qingwei Wen
- Pharmaceutical College, Guangxi Medical University, Guangxi, China
| | - Zhaoni Chen
- Pharmaceutical College, Guangxi Medical University, Guangxi, China
| | - Jinbin Wei
- Pharmaceutical College, Guangxi Medical University, Guangxi, China
| | - Tao Liang
- Pharmaceutical College, Guangxi Medical University, Guangxi, China
| | - Renbin Huang
- Pharmaceutical College, Guangxi Medical University, Guangxi, China
| |
Collapse
|
14
|
|
15
|
Zhang L, Zhao Y, Guo L. 17β-estradiol protects INS-1 insulinoma cells from mitophagy via G protein-coupled estrogen receptors and the PI3K/Akt signaling pathway. Int J Mol Med 2018; 41:2839-2846. [PMID: 29436590 DOI: 10.3892/ijmm.2018.3470] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 12/29/2017] [Indexed: 11/06/2022] Open
Abstract
17β-estradiol (17β-E2) is a steroid hormone that is known to exert effects on blood glucose homeostasis. The G protein‑coupled estrogen receptor (GPER) has been identified as a non-genomic estrogenic receptor, and is involved in numerous physiological processes, including cell survival, energy provision and metabolism. 17β-E2 may decrease apoptosis by binding to the GPER. The phosphoinositide 3-kinase (PI3K)/Akt signaling pathway is involved in physiological and pathological functions such as autophagy. The purpose of the present study was to investigate the role of the PI3K/Akt signaling pathway in the mediation of the effects of GPERs, and the effects of 17β-E2 on mitophagy in INS-1 cells, a rat insulin‑secreting β-cell line. In vitro, INS-1 cells were treated with different concentrations of 17β-E2 with and without pretreatment with a GPER antagonist (G15) or PI3K antagonist (LY294002) and compared with a negative control. An immunofluorescence assay demonstrated that GPERs are expressed in INS-1 cells. Western blot assays demonstrated that 17β-E2 increased GPER levels and the phosphorylation of Akt. Transmission electronic microscopy revealed that 17β-E2 reduced the formation of mitophagosomes and autophagosomes in INS-1 cells. An immunofluorescence staining assay indicated that the co-localization of translocase of mitochondrial outer membrane complex 20 (TOM20) with lysosomal-associated membrane protein 2 (LAMP2) was decreased in INS-1 cells treated with 17β-E2 alone. Western blotting demonstrated that 17β-E2 reduced the protein levels of activated microtubule-associated protein-1 light chain 3, and increased those of TOM20 and mitochondrial heat-shock protein 60. Notably, the protective effects of 17β-E2 were significantly diminished by G15 or LY294002. In conclusion, the present study suggests that 17β-E2 activates the PI3K/Akt pathway via the GPER in INS-1 cells. Furthermore, 17β-E2 may be involved in mitophagy by the regulating the GPER/PI3K/Akt pathway.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Endocrinology, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yuyan Zhao
- Department of Endocrinology, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Lei Guo
- Department of Orthopedic Surgery, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
16
|
Rojas J, Bermudez V, Palmar J, Martínez MS, Olivar LC, Nava M, Tomey D, Rojas M, Salazar J, Garicano C, Velasco M. Pancreatic Beta Cell Death: Novel Potential Mechanisms in Diabetes Therapy. J Diabetes Res 2018; 2018:9601801. [PMID: 29670917 PMCID: PMC5836465 DOI: 10.1155/2018/9601801] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 11/15/2017] [Accepted: 12/06/2017] [Indexed: 02/07/2023] Open
Abstract
PURPOSE OF REVIEW Describing the diverse molecular mechanisms (particularly immunological) involved in the death of the pancreatic beta cell in type 1 and type 2 diabetes mellitus. RECENT FINDINGS Beta cell death is the final event in a series of mechanisms that, up to date, have not been entirely clarified; it represents the pathophysiological mechanism in the natural history of diabetes mellitus. These mechanisms are not limited to an apoptotic process only, which is characteristic of the immune-mediated insulitis in type 1 diabetes mellitus. They also include the action of proinflammatory cytokines, the production of reactive oxygen species, DNA fragmentation (typical of necroptosis in type 1 diabetic patients), excessive production of islet amyloid polypeptide with the consequent endoplasmic reticulum stress, disruption in autophagy mechanisms, and protein complex formation, such as the inflammasome, capable of increasing oxidative stress produced by mitochondrial damage. SUMMARY Necroptosis, autophagy, and pyroptosis are molecular mechanisms that modulate the survival of the pancreatic beta cell, demonstrating the importance of the immune system in glucolipotoxicity processes and the potential role for immunometabolism as another component of what once known as the "ominous octet."
Collapse
Affiliation(s)
- Joselyn Rojas
- Pulmonary and Critical Care Medicine Department, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Endocrine and Metabolic Research Center, University of Zulia, Maracaibo, Venezuela
| | - Valmore Bermudez
- Endocrine and Metabolic Research Center, University of Zulia, Maracaibo, Venezuela
- Grupo de Investigación Altos Estudios de Frontera (ALEF), Universidad Simón Bolívar, Cúcuta, Colombia
| | - Jim Palmar
- Endocrine and Metabolic Research Center, University of Zulia, Maracaibo, Venezuela
| | - María Sofía Martínez
- Endocrine and Metabolic Research Center, University of Zulia, Maracaibo, Venezuela
| | - Luis Carlos Olivar
- Endocrine and Metabolic Research Center, University of Zulia, Maracaibo, Venezuela
| | - Manuel Nava
- Endocrine and Metabolic Research Center, University of Zulia, Maracaibo, Venezuela
| | - Daniel Tomey
- Endocrine and Metabolic Research Center, University of Zulia, Maracaibo, Venezuela
| | - Milagros Rojas
- Endocrine and Metabolic Research Center, University of Zulia, Maracaibo, Venezuela
| | - Juan Salazar
- Endocrine and Metabolic Research Center, University of Zulia, Maracaibo, Venezuela
| | - Carlos Garicano
- Grupo de Investigación Altos Estudios de Frontera (ALEF), Universidad Simón Bolívar, Cúcuta, Colombia
| | - Manuel Velasco
- Clinical Pharmacology Unit. School of Medicine José María Vargas, Central University of Venezuela, Caracas, Venezuela
| |
Collapse
|
17
|
Shpakov AO. [Pharmacological approaches for correction of thyroid dysfunctions in diabetes mellitus]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2017; 63:219-231. [PMID: 28781255 DOI: 10.18097/pbmc20176303219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Thyroid diseases are closely associated with the development of types 1 and 2 diabetes mellitus (DM), and as a consequence, the development of effective approaches for their treatment is one of the urgent problems of endocrinology. Traditionally, thyroid hormones (TH) are used to correct functions of the thyroid system. However, they are characterized by many side effects, such as their negative effect on the cardiovascular system as well as the ability of TH to enhance insulin resistance and to disturb insulin-producing function of pancreas, exacerbating thereby diabetic pathology. Therefore, the analogues of TH, selective for certain types of TH receptors, that do not have these side effects, are being developed. The peptide and low-molecular weight regulators of thyroid-stimulating hormone receptor, which regulate the activity of the thyroid axis at the stage of TH synthesis and secretion in thyrocytes, are being created. Systemic and intranasal administration of insulin, metformin therapy and drugs with antioxidant activity are effective for the treatment of thyroid pathology in types 1 and 2 DM. In the review, the literature data and the results of own investigations on pharmacological approaches for the treatment and prevention of thyroid diseases in patients with types 1 and 2 DM are summarized and analyzed.
Collapse
Affiliation(s)
- A O Shpakov
- I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences
| |
Collapse
|
18
|
Marchetti P, Bugliani M, De Tata V, Suleiman M, Marselli L. Pancreatic Beta Cell Identity in Humans and the Role of Type 2 Diabetes. Front Cell Dev Biol 2017; 5:55. [PMID: 28589121 PMCID: PMC5440564 DOI: 10.3389/fcell.2017.00055] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 05/05/2017] [Indexed: 12/13/2022] Open
Abstract
Pancreatic beta cells uniquely synthetize, store, and release insulin. Specific molecular, functional as well as ultrastructural traits characterize their insulin secretion properties and survival phentoype. In this review we focus on human islet/beta cells, and describe the changes that occur in type 2 diabetes and could play roles in the disease as well as represent possible targets for therapeutical interventions. These include transcription factors, molecules involved in glucose metabolism and insulin granule handling. Quantitative and qualitative insulin release patterns and their changes in type 2 diabetes are also associated with ultrastructural features involving the insulin granules, the mitochondria, and the endoplasmic reticulum.
Collapse
Affiliation(s)
- Piero Marchetti
- Department of Clinical and Experimental Medicine, University of PisaPisa, Italy
| | - Marco Bugliani
- Department of Clinical and Experimental Medicine, University of PisaPisa, Italy
| | - Vincenzo De Tata
- Department of Translational Medicine, University of PisaPisa, Italy
| | - Mara Suleiman
- Department of Clinical and Experimental Medicine, University of PisaPisa, Italy
| | - Lorella Marselli
- Department of Clinical and Experimental Medicine, University of PisaPisa, Italy
| |
Collapse
|
19
|
Shcherbina L, Edlund A, Esguerra JLS, Abels M, Zhou Y, Ottosson-Laakso E, Wollheim CB, Hansson O, Eliasson L, Wierup N. Endogenous beta-cell CART regulates insulin secretion and transcription of beta-cell genes. Mol Cell Endocrinol 2017; 447:52-60. [PMID: 28237718 DOI: 10.1016/j.mce.2017.02.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 02/16/2017] [Accepted: 02/16/2017] [Indexed: 01/20/2023]
Abstract
Impaired beta-cell function is key to the development of type 2 diabetes. Cocaine- and amphetamine-regulated transcript (CART) is an islet peptide with insulinotropic and glucagonostatic properties. Here we studied the role of endogenous CART in beta-cell function. CART silencing in INS-1 (832/13) beta-cells reduced insulin secretion and production, ATP levels and beta-cell exocytosis. This was substantiated by reduced expression of several exocytosis genes, as well as reduced expression of genes important for insulin secretion and processing. In addition, CART silencing reduced the expression of a network of transcription factors essential for beta-cell function. Moreover, in RNAseq data from human islet donors, CARTPT expression levels correlated with insulin, exocytosis genes and key beta-cell transcription factors. Thus, endogenous beta-cell CART regulates insulin expression and secretion in INS-1 (832/13) cells, via actions on the exocytotic machinery and a network of beta-cell transcription factors. We conclude that CART is important for maintaining the beta-cell phenotype.
Collapse
Affiliation(s)
- L Shcherbina
- Lund University Diabetes Centre, Skåne University Hospital, Jan Waldenströms Gata 35, 214 28 Malmö, Sweden
| | - A Edlund
- Lund University Diabetes Centre, Skåne University Hospital, Jan Waldenströms Gata 35, 214 28 Malmö, Sweden
| | - J L S Esguerra
- Lund University Diabetes Centre, Skåne University Hospital, Jan Waldenströms Gata 35, 214 28 Malmö, Sweden
| | - M Abels
- Lund University Diabetes Centre, Skåne University Hospital, Jan Waldenströms Gata 35, 214 28 Malmö, Sweden
| | - Y Zhou
- Lund University Diabetes Centre, Skåne University Hospital, Jan Waldenströms Gata 35, 214 28 Malmö, Sweden
| | - E Ottosson-Laakso
- Lund University Diabetes Centre, Skåne University Hospital, Jan Waldenströms Gata 35, 214 28 Malmö, Sweden
| | - C B Wollheim
- Lund University Diabetes Centre, Skåne University Hospital, Jan Waldenströms Gata 35, 214 28 Malmö, Sweden; Department of Cell Physiology and Metabolism, University Medical Center, 1 Rue Michel-Servet, CH-1211 Genève 4, Switzerland
| | - O Hansson
- Lund University Diabetes Centre, Skåne University Hospital, Jan Waldenströms Gata 35, 214 28 Malmö, Sweden
| | - L Eliasson
- Lund University Diabetes Centre, Skåne University Hospital, Jan Waldenströms Gata 35, 214 28 Malmö, Sweden
| | - N Wierup
- Lund University Diabetes Centre, Skåne University Hospital, Jan Waldenströms Gata 35, 214 28 Malmö, Sweden.
| |
Collapse
|
20
|
Fernandez-Gomez B, Ramos S, Goya L, Mesa MD, del Castillo MD, Martín MÁ. Coffee silverskin extract improves glucose-stimulated insulin secretion and protects against streptozotocin-induced damage in pancreatic INS-1E beta cells. Food Res Int 2016. [DOI: 10.1016/j.foodres.2016.03.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
21
|
Yang Z, Liu F, Qu H, Wang H, Xiao X, Deng H. 1, 25(OH)2D3 protects β cell against high glucose-induced apoptosis through mTOR suppressing. Mol Cell Endocrinol 2015. [PMID: 26213322 DOI: 10.1016/j.mce.2015.07.023] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Diabetes mellitus is a leading cause of death and disability worldwide, which presents a serious public health crisis in China nowadays. It has been well recognized that excessive β-cell apoptosis is the key pathogenesis of diabetes, of which the mammalian target of rapamycin (mTOR) serves as the critical signaling pathway. Emerging evidence indicates that vitamin D deficiency acts as a potential risk factor for diabetes. The present study aims to test the hypothesis that 1 alpha, 25-dihydroxyvitamin D(3) [1, 25(OH)2D3] can inhibit β-cell apoptosis via the suppression of mTOR signaling pathway. β-cells (INS-1) were cultured in the context of normal glucose or high glucose media with or without 1, 25(OH)2D3 treatment. β-cell apoptosis was evaluated by inverted fluorescence microscope, flow cytometry and electron microscope, respectively. Quantitative RT-PCR and Western blotting were performed to assess the possible perturbations in mTOR signaling pathway. High glucose significantly increased β-cell apoptosis. Of importance, RT-PCR and Western blotting demonstrated that high glucose inhibited DNA-damage-inducible transcript 4 (DDIT4) and TSC1/TSC2, up-regulated Rheb/mTOR/p70S6K and enhanced expression of the apoptosis regulating proteins, such as phospho-Bcl-2, cytochrome C and cleaved caspase. Interestingly, 1, 25(OH)2D3 treatment reversed high glucose induced pathological changes in mTOR signaling pathway, restored expression of DDIT4 and TSC1/TSC2, blocked aberrant up-regulation of Rheb/mTOR/p70S6K and the apoptosis regulating proteins, and effectively inhibited β-cell apoptosis. Therefore, 1, 25(OH)2D3 treatment can effectively protects β cell against high glucose-induced apoptosis mainly via the suppression of mTOR signaling pathway, which may be considered as a potential therapy for patients with diabetes.
Collapse
Affiliation(s)
- Zesong Yang
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, PR China
| | - Fang Liu
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, PR China
| | - Hua Qu
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, PR China
| | - Hang Wang
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, PR China
| | - Xiaoqiu Xiao
- Laboratory of Lipid and Glucose Metabolism, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, PR China
| | - Huacong Deng
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, PR China.
| |
Collapse
|
22
|
Song Y, Wan X, Gao L, Pan Y, Xie W, Wang H, Guo J. Activated PKR inhibits pancreatic β-cell proliferation through sumoylation-dependent stabilization of P53. Mol Immunol 2015; 68:341-9. [PMID: 26446704 DOI: 10.1016/j.molimm.2015.09.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 08/27/2015] [Accepted: 09/02/2015] [Indexed: 12/20/2022]
Abstract
Double-stranded RNA-dependent protein kinase (PKR) is intimately involved in type 2 diabetes due to its role in insulin resistance in peripheral tissues and anti-proliferative effect on pancreatic β-cells. Activated PKR was found to inhibit β-cell proliferation, partially through accumulation of P53. However the molecular mechanisms underlying PKR-dependent upregulation of P53 remain unknown. The results of the present study showed that PKR can be specifically activated in PKR overexpressing β-cells by a low dosage of the previously synthesized compound 1H-benzimidazole1-ethanol,2,3-dihydro-2-imino-a-(phenoxymethyl)-3-(phenylmethyl)-,monohydrochloride (BEPP), and this led to upregulation of P53 through sumoylation-dependent stability. Activated PKR was found to interact with sumo-conjugating enzyme Ubc9, and P53 sumoylation relies on a PKR-Ubc9 protein-protein interaction. Additionally, a ceramide signal was needed for PKR activation to be triggered by glucolipotoxicity and TNFα stimulation, and stabilization of P53 required endogenous ceramide accumulation. Glucolipotoxicity and pro-inflammatory cytokines therefore promote the sumoylation-dependent stability of P53 via the ceramide/PKR/Ubc9 signalling pathway that is involved in pancreatic β-cell proliferation inhibition in the development of type 2 diabetes.
Collapse
Affiliation(s)
- Ying Song
- Department of Respiratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - XiaoMeng Wan
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing 210029, PR China
| | - LiLi Gao
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing 210029, PR China
| | - Yi Pan
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing 210029, PR China
| | - WeiPing Xie
- Department of Respiratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Hong Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China.
| | - Jun Guo
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing 210029, PR China
| |
Collapse
|
23
|
Yao XF, Zheng BL, Bai J, Jiang LP, Zheng Y, Qi BX, Geng CY, Zhong LF, Yang G, Chen M, Liu XF, Sun XC. Low-level sodium arsenite induces apoptosis through inhibiting TrxR activity in pancreatic β-cells. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2015; 40:486-91. [PMID: 26291581 DOI: 10.1016/j.etap.2015.08.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 08/03/2015] [Accepted: 08/05/2015] [Indexed: 05/02/2023]
Abstract
In our previous study, we reported that sodium arsenite induced ROS-dependent apoptosis through lysosomal-mitochondrial pathway in pancreatic β-cells. Since the thioredoxin (Trx) system is the key antioxidant factor in mammalian cells, we investigate whether the inhibition of Trx system contributes to sodium arsenite-induced apoptosis in this study. After treatment with low-level (0.25-1μM) sodium arsenite for 96h, the thioredoxin reductase (TrxR) activity was decreased significantly in pancreatic INS-1 cells. Following with the inactivation of TrxR, ASK1 was released from combining with Trx, which was evidenced by increased levels of ASK1 in sodium arsenite-treated INS-1 cells. Subsequently, activated ASK1 accelerated the expression of proapoptotic protein Bax and reduced the expression of anti-apoptic protein Bcl-2. Finally, low-level sodium arsenite induced apoptosis via caspase-3 in INS-1 cells. Knockdown of ASK1 alleviated sodium arsenite-induced apoptosis. In summary, the precise molecular mechanisms through which arsenic is related to diabetes have not been completely elucidated, inactivation of Trx system might provide insights into the underlying mechanisms at the environmental exposure levels.
Collapse
Affiliation(s)
- Xiao-Feng Yao
- Department of Occupational and Environmental Health, Dalian Medical University, 9 W Lvshun South Road, Dalian 116044, PR China
| | - Bai-Lu Zheng
- Department of Occupational and Environmental Health, Dalian Medical University, 9 W Lvshun South Road, Dalian 116044, PR China
| | - Jie Bai
- Department of Occupational and Environmental Health, Dalian Medical University, 9 W Lvshun South Road, Dalian 116044, PR China
| | - Li-Ping Jiang
- Liaoning Anti-Degenerative Diseases Natural Products Engineering Research Center, Dalian Medical University, 9 W Lvshun South Road, Dalian 116044, PR China
| | - Yue Zheng
- Department of Occupational and Environmental Health, Dalian Medical University, 9 W Lvshun South Road, Dalian 116044, PR China
| | - Bao-Xu Qi
- Department of Occupational and Environmental Health, Dalian Medical University, 9 W Lvshun South Road, Dalian 116044, PR China
| | - Cheng-Yan Geng
- Liaoning Anti-Degenerative Diseases Natural Products Engineering Research Center, Dalian Medical University, 9 W Lvshun South Road, Dalian 116044, PR China
| | - Lai-Fu Zhong
- Liaoning Anti-Degenerative Diseases Natural Products Engineering Research Center, Dalian Medical University, 9 W Lvshun South Road, Dalian 116044, PR China
| | - Guang Yang
- Liaoning Anti-Degenerative Diseases Natural Products Engineering Research Center, Dalian Medical University, 9 W Lvshun South Road, Dalian 116044, PR China
| | - Min Chen
- Department of Occupational and Environmental Health, Dalian Medical University, 9 W Lvshun South Road, Dalian 116044, PR China
| | - Xiao-Fang Liu
- Department of Occupational and Environmental Health, Dalian Medical University, 9 W Lvshun South Road, Dalian 116044, PR China
| | - Xian-Ce Sun
- Department of Occupational and Environmental Health, Dalian Medical University, 9 W Lvshun South Road, Dalian 116044, PR China; Liaoning Anti-Degenerative Diseases Natural Products Engineering Research Center, Dalian Medical University, 9 W Lvshun South Road, Dalian 116044, PR China.
| |
Collapse
|
24
|
Rodríguez-Castelán J, Nicolás L, Morimoto S, Cuevas E. The Langerhans islet cells of female rabbits are differentially affected by hypothyroidism depending on the islet size. Endocrine 2015; 48:811-7. [PMID: 25213470 DOI: 10.1007/s12020-014-0418-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 09/01/2014] [Indexed: 12/31/2022]
Abstract
Effects of hypothyroidism on the glucose and insulin levels are controversial, and its impact on the Langerhans islet morphology of adult subjects has been poorly addressed. In spite of hypothyroidism and diabetes mellitus are more frequent in females than in males, most studies using animal models have been done in males. The effect of hypothyroidism on the immunolabeling of thyroid hormone receptors (TRs) and thyrotropin receptor (TSHR) of islet cells is unknown. The aim of this study was to determine the effect of hypothyroidism on the glucose and insulin concentrations, morphometry of islets, and immunostaining of TRs α1-2 and β1 and TSHR of islet cells in female rabbits. Control and hypothyroid (0.02% of methimazole for 30 days) animals were used to quantify blood levels of glucose and insulin, density of islets, cross-sectional area (CSA) of islets, number of cells per islet, cell proliferation, and the immunolabeling of TRs α1-2, TRβ1, and TSHR. Student's t or Mann-Whitney-U tests, two-way ANOVAs, and Fischer's tests were applied. Concentrations of glucose and insulin, as well as the insulin resistance were similar between groups. Hypothyroidism did not affect the density or the CSA of islets. The analysis of islets by size showed that hypothyroidism reduced the cell number in large and medium islets, but not in small ones. In small islets, cell proliferation was increased. The immunoreactivity of TRα1-2, TRβ1, and TSHR was increased by hypothyroidism in all islet sizes. Our results show that hypothyroidism affects differentially the islet cells depending on the size of islets.
Collapse
Affiliation(s)
- J Rodríguez-Castelán
- Maestría en Ciencias Biológicas, Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico
| | | | | | | |
Collapse
|
25
|
Campos C. Chronic Hyperglycemia and Glucose Toxicity: Pathology and Clinical Sequelae. Postgrad Med 2015; 124:90-7. [DOI: 10.3810/pgm.2012.11.2615] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
26
|
Qi Nan W, Ling Z, Bing C. The influence of the telomere-telomerase system on diabetes mellitus and its vascular complications. Expert Opin Ther Targets 2015; 19:849-64. [PMID: 25677239 DOI: 10.1517/14728222.2015.1016500] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION The telomere-telomerase system plays an important role in the pathogenesis and disease progression of diabetes mellitus as well as in its vascular complications. Recent studies suggest that telomere shortening and abnormal telomerase activity occur in patients with diabetes mellitus, and targeting the telomere-telomerase system has become a prospective treatment for diabetes mellitus and its vascular complications. This review highlights the significance of the telomere-telomerase system and supports its role as a possible therapeutic target for patients with diabetes mellitus and its vascular complications Areas covered: This review covers the advances in understanding the telomere-telomerase system over the last 30 years and its significance in diabetes mellitus. In addition, it provides knowledge regarding the significance of the telomere-telomerase system in diabetes mellitus and its vascular complications as well as its role and mechanisms in oxidative stress, cell therapy and antioxidant activity Expert opinion: The telomere-telomerase system may be a potential therapeutic target that can protect against DNA damage and apoptosis in patients with diabetes mellitus and its vascular complications. DNA damage and apoptosis are associated with oxidative stress and are involved in the dysfunction of pancreatic β cells, insulin resistance, and its vascular complications. Abnormalities in the telomere-telomerase system may be associated with diabetes mellitus and its vascular complications. Therapies targeting telomere-telomerase system, telomerase reverse transcriptase transfection and alterative telomere lengthening must be identified before gene therapy can commence.
Collapse
Affiliation(s)
- Wu Qi Nan
- The First Affiliated Hospital of the Third Military Medical University, Endocrine Department , Chongqing, Post number: 400038 , China
| | | | | |
Collapse
|
27
|
Jiang N, Zhang S, Zhu J, Shang J, Gao X. Hypoglycemic, Hypolipidemic and Antioxidant Effects of Peptides from Red Deer Antlers in Streptozotocin-Induced Diabetic Mice. TOHOKU J EXP MED 2015; 236:71-9. [DOI: 10.1620/tjem.236.71] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Ning Jiang
- Department of Biological Science and Technology, Hubei University for Nationalities
- School of Life Science and Technology, China Pharmaceutical University
| | - Shuangjian Zhang
- School of Life Science and Technology, China Pharmaceutical University
| | - Jing Zhu
- School of Life Science and Technology, China Pharmaceutical University
| | - Jing Shang
- New Drug Screening Center, China Pharmaceutical University
| | - Xiangdong Gao
- School of Life Science and Technology, China Pharmaceutical University
| |
Collapse
|
28
|
Chung JO, Cho DH, Chung DJ, Chung MY. Serum bilirubin concentrations are positively associated with serum C-peptide levels in patients with Type 2 diabetes. Diabet Med 2014; 31:1316-22. [PMID: 24910105 DOI: 10.1111/dme.12516] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/03/2014] [Indexed: 01/02/2023]
Abstract
AIMS To investigate the relationship between physiological serum total bilirubin concentrations and serum C-peptide levels in Korean patients with Type 2 diabetes. METHODS A total of 588 patients with Type 2 diabetes were investigated in this cross-sectional study. Fasting C-peptide level, 2-h postprandial C-peptide level and ΔC-peptide (postprandial C-peptide minus fasting C-peptide) level were measured in all patients. RESULTS Fasting C-peptide level, postprandial C-peptide level and ΔC-peptide level tended to be higher in patients with higher bilirubin concentrations. Partial correlation analysis showed that serum bilirubin levels were significantly correlated with fasting C-peptide level (r = 0.159, P < 0.001), postprandial C-peptide level (r = 0.209, P < 0.001) and ΔC-peptide level (r = 0.186, P < 0.001) after adjustment for other covariates. In the multivariate model, the association between serum bilirubin concentrations and serum C-peptide levels remained significant after adjustment for confounding factors including age, gender, familial diabetes, hypertension, hyperlipidaemia, BMI, HbA1c , duration of diabetes and associated liver function tests (fasting C-peptide level: β = 0.083, P = 0.041; postprandial C-peptide level: β = 0.106, P = 0.005; ΔC-peptide level: β = 0.096, P = 0.015, respectively). CONCLUSIONS Serum bilirubin concentrations within the physiological range were positively associated with serum C-peptide levels in patients with Type 2 diabetes.
Collapse
Affiliation(s)
- J O Chung
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea
| | | | | | | |
Collapse
|
29
|
Inagaki N, Kondo K, Yoshinari T, Kuki H. Efficacy and safety of canagliflozin alone or as add-on to other oral antihyperglycemic drugs in Japanese patients with type 2 diabetes: A 52-week open-label study. J Diabetes Investig 2014; 6:210-8. [PMID: 25802729 PMCID: PMC4364856 DOI: 10.1111/jdi.12266] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 07/04/2014] [Accepted: 07/07/2014] [Indexed: 01/09/2023] Open
Abstract
Aims/Introduction Canagliflozin is a sodium–glucose cotransporter 2 inhibitor under development for the treatment of type 2 diabetes. Our aim was to examine its efficacy and safety as monotherapy or in combination with commonly used oral antihyperglycemic drugs in Japanese patients with type 2 diabetes. Materials and Methods Patients on diet/exercise alone or diet/exercise plus an oral antihyperglycemic drug (sulfonylurea, glinide, α-glucosidase inhibitor, biguanide, thiazolidinedione or dipeptidyl peptidase-4 inhibitor) were randomized to either 100 or 200 mg canagliflozin while continuing prior therapy. Patients were treated for 52 weeks in an open-label manner. Results Canagliflozin significantly reduced hemoglobin A1c, fasting plasma glucose and bodyweight in all the study groups. Improvements were apparent by 4 weeks of treatment, and were maintained for 52 weeks. The reduction in hemoglobin A1c ranged from −0.80 to −1.06%, and from −0.93 to −1.26% in the 100 and 200 mg canagliflozin groups, respectively. Drug-related adverse events occurred in approximately one-third of patients, and included hypoglycemia/asymptomatic hypoglycemia and pollakiuria. Hypoglycemia/asymptomatic hypoglycemia was most common in patients treated with a sulfonylurea. Most adverse events were classified as mild or moderate in severity. Conclusions The results of the present study confirmed that treatment with canagliflozin resulted in significant reductions in glycemic control and bodyweight that were maintained for 52 weeks of treatment irrespective of whether it was administered as monotherapy or in combination with another oral antihyperglycemic drug. Canagliflozin was well tolerated, with a low incidence of drug-related adverse events. This trial was registered with ClinicalTrials.gov (no. NCT01387737).
Collapse
Affiliation(s)
- Nobuya Inagaki
- Department of Diabetes Endocrinology and Nutrition, Kyoto University Graduate School of Medicine Kyoto, Japan
| | | | | | - Hideki Kuki
- Mitsubishi Tanabe Pharma Corporation Tokyo, Japan
| |
Collapse
|
30
|
Biden TJ, Boslem E, Chu KY, Sue N. Lipotoxic endoplasmic reticulum stress, β cell failure, and type 2 diabetes mellitus. Trends Endocrinol Metab 2014; 25:389-98. [PMID: 24656915 DOI: 10.1016/j.tem.2014.02.003] [Citation(s) in RCA: 165] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Revised: 02/12/2014] [Accepted: 02/19/2014] [Indexed: 02/06/2023]
Abstract
Failure of the unfolded protein response (UPR) to maintain optimal folding of pro-insulin in the endoplasmic reticulum (ER) leads to unresolved ER stress and β cell death. This contributes not only to some rare forms of diabetes, but also to type 2 diabetes mellitus (T2DM). Many key findings, elaborated over the past decade, are based on the lipotoxicity model, entailing chronic exposure of β cells to elevated levels of fatty acids (FAs). Here, we update recent progress on how FAs initiate ER stress, particularly via disruption of protein trafficking, and how this leads to apoptosis. We also highlight differences in how β cells are impacted by the classic UPR, versus the more selective UPR that arises as part of a broader response to lipotoxicity.
Collapse
Affiliation(s)
- Trevor J Biden
- Diabetes and Obesity Program, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia.
| | - Ebru Boslem
- Diabetes and Obesity Program, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Kwan Yi Chu
- Diabetes and Obesity Program, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Nancy Sue
- Diabetes and Obesity Program, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| |
Collapse
|
31
|
Scharp DW, Marchetti P. Encapsulated islets for diabetes therapy: history, current progress, and critical issues requiring solution. Adv Drug Deliv Rev 2014; 67-68:35-73. [PMID: 23916992 DOI: 10.1016/j.addr.2013.07.018] [Citation(s) in RCA: 214] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 07/10/2013] [Accepted: 07/22/2013] [Indexed: 02/07/2023]
Abstract
Insulin therapy became a reality in 1921 dramatically saving lives of people with diabetes, but not protecting them from long-term complications. Clinically successful free islet implants began in 1989 but require life long immunosuppression. Several encapsulated islet approaches have been ongoing for over 30 years without defining a clinically relevant product. Macro-devices encapsulating islet mass in a single device have shown long-term success in large animals but human trials have been limited by critical challenges. Micro-capsules using alginate or similar hydrogels encapsulate individual islets with many hundreds of promising rodent results published, but a low incidence of successful translation to large animal and human results. Reduction of encapsulated islet mass for clinical transplantation is in progress. This review covers the status of both early and current studies including the presentation of corporate efforts involved. It concludes by defining the critical items requiring solution to enable a successful clinical diabetes therapy.
Collapse
|
32
|
Fernández-Millán E, Ramos S, Alvarez C, Bravo L, Goya L, Martín MÁ. Microbial phenolic metabolites improve glucose-stimulated insulin secretion and protect pancreatic beta cells against tert-butyl hydroperoxide-induced toxicity via ERKs and PKC pathways. Food Chem Toxicol 2014; 66:245-53. [PMID: 24491264 DOI: 10.1016/j.fct.2014.01.044] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 01/24/2014] [Accepted: 01/27/2014] [Indexed: 12/26/2022]
Abstract
Oxidative stress is accepted as one of the causes of beta cell failure in type 2 diabetes. Therefore, identification of natural antioxidant agents that preserve beta cell mass and function is considered an interesting strategy to prevent or treat diabetes. Recent evidences indicated that colonic metabolites derived from flavonoids could possess beneficial effects on various tissues. The aim of this work was to establish the potential anti-diabetic properties of the microbial-derived flavonoid metabolites 3,4-dihydroxyphenylacetic acid (DHPAA), 2,3-dihydroxybenzoic acid (DHBA) and 3-hydroxyphenylpropionic acid (HPPA). To this end, we tested their ability to influence beta cell function and to protect against tert-butyl hydroperoxide-induced beta cell toxicity. DHPAA and HPPA were able to potentiate glucose-stimulated insulin secretion (GSIS) in a beta cell line INS-1E and in rat pancreatic islets. Moreover, pre-treatment of cells with both compounds protected against beta cell dysfunction and death induced by the pro-oxidant. Finally, experiments with pharmacological inhibitors indicate that these effects were mediated by the activation of protein kinase C and the extracellular regulated kinases pathways. Altogether, these findings strongly suggest that the microbial-derived flavonoid metabolites DHPAA and HPPA may have anti-diabetic potential by promoting survival and function of pancreatic beta cells.
Collapse
Affiliation(s)
- Elisa Fernández-Millán
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Sonia Ramos
- Departamento de Metabolismo y Nutrición, Instituto de Ciencia y Tecnología de los Alimentos y Nutrición (ICTAN), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Carmen Alvarez
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain; Departamento de Bioquímica y Biología Molecular II, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Laura Bravo
- Departamento de Metabolismo y Nutrición, Instituto de Ciencia y Tecnología de los Alimentos y Nutrición (ICTAN), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Luis Goya
- Departamento de Metabolismo y Nutrición, Instituto de Ciencia y Tecnología de los Alimentos y Nutrición (ICTAN), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - María Ángeles Martín
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain; Departamento de Metabolismo y Nutrición, Instituto de Ciencia y Tecnología de los Alimentos y Nutrición (ICTAN), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.
| |
Collapse
|
33
|
Iwata M, Matsushita Y, Fukuda K, Wakura T, Okabe K, Koshimizu Y, Fukushima Y, Kobashi C, Yamazaki Y, Honoki H, Suzuki H, Kigawa M, Tobe K. Secretory units of islets in transplantation index is a useful predictor of insulin requirement in Japanese type 2 diabetic patients. J Diabetes Investig 2013; 5:570-80. [PMID: 25411626 PMCID: PMC4188116 DOI: 10.1111/jdi.12181] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 10/18/2013] [Accepted: 10/22/2013] [Indexed: 11/28/2022] Open
Abstract
AIMS/INTRODUCTION The objective of the present study was to clarify the validity of β-cell function-related parameters for predicting the insulin requirement of Japanese type 2 diabetic patients. MATERIALS AND METHODS In 188 patients with type 2 diabetes who had been admitted to the University of Toyama Hospital (Toyama, Japan) without receiving insulin therapy, we carried out a cross-sectional study examining the relationship between the homeostasis model assessment of β-cell function (HOMA-β) and C-peptide-based indices, and also carried out a retrospective study to examine the utility for predicting insulin requirement of several β -cell function-related indices using a receiver operating characteristic (ROC) curve analysis. RESULTS The secretory units of islets in transplantation index (SUIT) had the strongest correlation with HOMA-β, followed by the fasting serum C-peptide immunoreactivity index (CPI); the fasting serum C-peptide immunoreactivity itself (F-CPR) had the least correlation. The CPI, HOMA-β and SUIT were significantly lower in the insulin-requiring group than in the non-insulin-requiring group, even after adjustments for confounding factors (P < 0.01). The areas under the ROC curve for insulin requirement were 0.622, 0.774, 0.808, and 0.759 for F-CPR, CPI, SUIT, and HOMA-β, respectively. The cut-off values of SUIT, CPI, and HOMA-β for an over 80% specificity for the prediction of insulin therapy were 23.5, 1.00, and 14.9, respectively. CONCLUSIONS The present study shows that SUIT is the best predictor of insulin requirement among these β-cell function-related markers.
Collapse
Affiliation(s)
- Minoru Iwata
- First Department of Internal Medicine University of Toyama Toyama Japan ; Community Medical Support Unit Faculty of Medicine University of Toyama Toyama Japan
| | - Yumi Matsushita
- Department of Clinical Research National Center for Global Health and Medicine Tokyo Japan
| | - Kazuhito Fukuda
- First Department of Internal Medicine University of Toyama Toyama Japan
| | - Tatsurou Wakura
- First Department of Internal Medicine University of Toyama Toyama Japan
| | - Keisuke Okabe
- First Department of Internal Medicine University of Toyama Toyama Japan
| | - Yukiko Koshimizu
- First Department of Internal Medicine University of Toyama Toyama Japan
| | - Yasuo Fukushima
- Department of Internal Medicine Asahi General Hospital Asahi-machi Japan
| | - Chikaaki Kobashi
- Department of Internal Medicine Kamiichi General Hospital Kamiichi-machi Japan
| | - Yu Yamazaki
- Department of Internal Medicine Saiseikai Toyama Hospital Toyama Japan
| | - Hisae Honoki
- Division of Endocrinology and Metabolism Department of Internal Medicine Saiseikai Takaoka Hospital Takaoka Toyama Japan
| | - Hikari Suzuki
- Department of Internal Medicine Shakaihoken Takaoka Hospital Takaoka Toyama Japan
| | - Mika Kigawa
- Department of Public Health University of Toyama Toyama Japan
| | - Kazuyuki Tobe
- First Department of Internal Medicine University of Toyama Toyama Japan
| |
Collapse
|
34
|
Patané G, Caporarello N, Marchetti P, Parrino C, Sudano D, Marselli L, Vigneri R, Frittitta L. Adiponectin increases glucose-induced insulin secretion through the activation of lipid oxidation. Acta Diabetol 2013; 50:851-7. [PMID: 23440352 DOI: 10.1007/s00592-013-0458-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Accepted: 01/28/2013] [Indexed: 01/08/2023]
Abstract
The expression of adiponectin receptors has been demonstrated in human and rat pancreatic beta cells, where globular (g) adiponectin rescues rat beta cells from cytokine and fatty acid-induced apoptosis. The aim of our study was to evaluate whether adiponectin has a direct effect on insulin secretion and the metabolic pathways involved. Purified human pancreatic islets and rat beta cells (INS-1E) were exposed (1 h) to g-adiponectin, and glucose-induced insulin secretion was measured. A significant increase in glucose-induced insulin secretion was observed in the presence of g-adiponectin (1 nmol/l) with respect to control cells in both human pancreatic islets (n = 5, p < 0.05) and INS-1E cells (n = 5, p < 0.001). The effect of globular adiponectin on insulin secretion was independent of AMP-dependent protein kinase (AMPK) activation or glucose oxidation. In contrast, g-adiponectin significantly increased oleate oxidation (n = 5, p < 0.05), and the effect of g-adiponectin (p < 0.001) on insulin secretion by INS-1E was significantly reduced in the presence of etomoxir (1 μmol/l), an inhibitor of fatty acid beta oxidation. g-Adiponectin potentiates glucose-induced insulin secretion in both human pancreatic islets and rat beta cells via an AMPK independent pathway. Increased fatty acid oxidation rather than augmented glucose oxidation is the mechanism responsible. Overall, our data indicate that, in addition to its anti-apoptotic action, g-adiponectin has another direct effect on beta cells by potentiating insulin secretion. Adiponectin, therefore, in addition to its well-known effect on insulin sensitivity, has important effects at the pancreatic level.
Collapse
Affiliation(s)
- G Patané
- Unit of Endocrinology, Department of Clinical and Molecular Biomedicine, Garibaldi-Nesima Hospital, University of Catania Medical School, Via Palermo 636, 95122, Catania, Italy
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Natalicchio A, Labarbuta R, Tortosa F, Biondi G, Marrano N, Peschechera A, Carchia E, Orlando MR, Leonardini A, Cignarelli A, Marchetti P, Perrini S, Laviola L, Giorgino F. Exendin-4 protects pancreatic beta cells from palmitate-induced apoptosis by interfering with GPR40 and the MKK4/7 stress kinase signalling pathway. Diabetologia 2013; 56:2456-66. [PMID: 23995397 DOI: 10.1007/s00125-013-3028-4] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2013] [Accepted: 07/29/2013] [Indexed: 12/24/2022]
Abstract
AIMS/HYPOTHESIS The mechanisms of the protective effects of exendin-4 on NEFA-induced beta cell apoptosis were investigated. METHODS The effects of exendin-4 and palmitate were evaluated in human and murine islets, rat insulin-secreting INS-1E cells and murine glucagon-secreting alpha-TC1-6 cells. mRNA and protein expression/phosphorylation were measured by real-time RT-PCR and immunoblotting or immunofluorescence, respectively. Small interfering (si)RNAs for Ib1 and Gpr40 were used. Cell apoptosis was quantified by two independent assays. Insulin release was assessed with an insulin ELISA. RESULTS Exposure of human and murine primary islets and INS-1E cells, but not alpha-TC1-6 cells, to exendin-4 inhibited phosphorylation of the stress kinases, c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK), and prevented apoptosis in response to palmitate. Exendin-4 increased the protein content of islet-brain 1 (IB1), an endogenous JNK blocker; however, siRNA-mediated reduction of IB1 did not impair the ability of exendin-4 to inhibit JNK and prevent apoptosis. Exendin-4 reduced G-protein-coupled receptor 40 (GPR40) expression and inhibited palmitate-induced phosphorylation of mitogen-activated kinase kinase (MKK)4 and MKK7. The effects of exendin-4 were abrogated in the presence of the protein kinase A (PKA) inhibitors, H89 and KT5720. Knockdown of GPR40, as well as use of a specific GPR40 antagonist, resulted in diminished palmitate-induced JNK and p38 MAPK phosphorylation and apoptosis. Furthermore, inhibition of JNK and p38 MAPK activity prevented palmitate-induced apoptosis. CONCLUSIONS/INTERPRETATION Exendin-4 counteracts the proapoptotic effects of palmitate in beta cells by reducing GPR40 expression and inhibiting MKK7- and MKK4-dependent phosphorylation of the stress kinases, JNK and p38 MAPK, in a PKA-dependent manner.
Collapse
Affiliation(s)
- Annalisa Natalicchio
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Pancreatic beta-cell mass expands through beta-cell proliferation and neogenesis while it decreases mainly via apoptosis. The loss of balance between beta-cell death and regeneration leads to a reduction of beta-cell functional mass, thus contributing to the pathogenesis of type 2 diabetes mellitus (T2DM). The pathogenetic mechanisms causing T2DM are complex, and also include a significant reduction of the incretin effect. A better understanding of the role of incretin hormones in glucose homeostasis has led to the development of incretin-based therapies. Recently, incretin hormones have been shown to stimulate the beta-cell growth and differentiation from pancreas-derived stem/progenitor cells, as well as to exert cytoprotective, antiapoptotic effects on beta-cells. However, the role and the molecular mechanisms by which GLP-1 and its agonists regulate beta-cell mass have not been fully investigated. This review focuses the current findings and the missing understanding of the effects of incretin hormones on beta-cell mass expansion.
Collapse
Affiliation(s)
- Federica Tortosa
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari School of Medicine , Bari , Italy and
| | | |
Collapse
|
37
|
Bugliani M, Liechti R, Cheon H, Suleiman M, Marselli L, Kirkpatrick C, Filipponi F, Boggi U, Xenarios I, Syed F, Ladriere L, Wollheim C, Lee MS, Marchetti P. Microarray analysis of isolated human islet transcriptome in type 2 diabetes and the role of the ubiquitin-proteasome system in pancreatic beta cell dysfunction. Mol Cell Endocrinol 2013; 367:1-10. [PMID: 23246353 DOI: 10.1016/j.mce.2012.12.001] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Revised: 12/04/2012] [Accepted: 12/04/2012] [Indexed: 12/14/2022]
Abstract
To shed light on islet cell molecular phenotype in human type 2 diabetes (T2D), we studied the transcriptome of non-diabetic (ND) and T2D islets to then focus on the ubiquitin-proteasome system (UPS), the major protein degradation pathway. We assessed gene expression, amount of ubiquitinated proteins, proteasome activity, and the effects of proteasome inhibition and prolonged exposure to palmitate. Microarray analysis identified more than one thousand genes differently expressed in T2D islets, involved in many structures and functions, with consistent alterations of the UPS. Quantitative RT-PCR demonstrated downregulation of selected UPS genes in T2D islets and beta cell fractions, with greater ubiquitin accumulation and reduced proteasome activity. Chemically induced reduction of proteasome activity was associated with lower glucose-stimulated insulin secretion, which was partly reproduced by palmitate exposure. These results show the presence of many changes in islet transcriptome in T2D islets and underline the importance of the association between UPS alterations and beta cell dysfunction in human T2D.
Collapse
Affiliation(s)
- Marco Bugliani
- Department of Clinical and Experimental Medicine, Islet Cell Laboratory, University of Pisa, Pisa 56124, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Sanlioglu AD, Altunbas HA, Balci MK, Griffith TS, Sanlioglu S. Clinical utility of insulin and insulin analogs. Islets 2013; 5:67-78. [PMID: 23584214 PMCID: PMC4204021 DOI: 10.4161/isl.24590] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 04/05/2013] [Accepted: 04/06/2013] [Indexed: 02/08/2023] Open
Abstract
Diabetes is a pandemic disease characterized by autoimmune, genetic and metabolic abnormalities. While insulin deficiency manifested as hyperglycemia is a common sequel of both Type-1 and Type-2 diabetes (T1DM and T2DM), it does not result from a single genetic defect--rather insulin deficiency results from the functional loss of pancreatic β cells due to multifactorial mechanisms. Since pancreatic β cells of patients with T1DM are destroyed by autoimmune reaction, these patients require daily insulin injections. Insulin resistance followed by β cell dysfunction and β cell loss is the characteristics of T2DM. Therefore, most patients with T2DM will require insulin treatment due to eventual loss of insulin secretion. Despite the evidence of early insulin treatment lowering macrovascular (coronary artery disease, peripheral arterial disease and stroke) and microvascular (diabetic nephropathy, neuropathy and retinopathy) complications of T2DM, controversy exists among physicians on how to initiate and intensify insulin therapy. The slow acting nature of regular human insulin makes its use ineffective in counteracting postprandial hyperglycemia. Instead, recombinant insulin analogs have been generated with a variable degree of specificity and action. Due to the metabolic variability among individuals, optimum blood glucose management is a formidable task to accomplish despite the presence of novel insulin analogs. In this article, we present a recent update on insulin analog structure and function with an overview of the evidence on the various insulin regimens clinically used to treat diabetes.
Collapse
MESH Headings
- Animals
- Diabetes Mellitus, Type 1/blood
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/drug therapy
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/metabolism
- Diabetic Angiopathies/prevention & control
- Drug Monitoring
- Evidence-Based Medicine
- Humans
- Hyperglycemia/prevention & control
- Hypoglycemic Agents/administration & dosage
- Hypoglycemic Agents/chemistry
- Hypoglycemic Agents/metabolism
- Hypoglycemic Agents/therapeutic use
- Insulin/administration & dosage
- Insulin/analogs & derivatives
- Insulin/metabolism
- Insulin/therapeutic use
- Insulin, Regular, Human/administration & dosage
- Insulin, Regular, Human/analogs & derivatives
- Insulin, Regular, Human/genetics
- Insulin, Regular, Human/therapeutic use
- Recombinant Proteins/administration & dosage
- Recombinant Proteins/chemistry
- Recombinant Proteins/therapeutic use
Collapse
Affiliation(s)
- Ahter D. Sanlioglu
- Human Gene and Cell Therapy Center; Akdeniz University Faculty of Medicine; Antalya, Turkey
- Department of Medical Biology and Genetics; Akdeniz University Faculty of Medicine; Antalya, Turkey
| | - Hasan Ali Altunbas
- Human Gene and Cell Therapy Center; Akdeniz University Faculty of Medicine; Antalya, Turkey
- Department of Internal Medicine; Division of Endocrinology and Metabolism; Akdeniz University Faculty of Medicine; Antalya, Turkey
| | - Mustafa Kemal Balci
- Human Gene and Cell Therapy Center; Akdeniz University Faculty of Medicine; Antalya, Turkey
- Department of Internal Medicine; Division of Endocrinology and Metabolism; Akdeniz University Faculty of Medicine; Antalya, Turkey
| | | | - Salih Sanlioglu
- Human Gene and Cell Therapy Center; Akdeniz University Faculty of Medicine; Antalya, Turkey
- Department of Medical Biology and Genetics; Akdeniz University Faculty of Medicine; Antalya, Turkey
| |
Collapse
|
39
|
Comley RA, Kallend D. Imaging in the cardiovascular and metabolic disease area. Drug Discov Today 2013; 18:185-92. [DOI: 10.1016/j.drudis.2012.09.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Revised: 09/14/2012] [Accepted: 09/24/2012] [Indexed: 01/09/2023]
|
40
|
Gallwitz B, Kazda C, Kraus P, Nicolay C, Schernthaner G. Contribution of insulin deficiency and insulin resistance to the development of type 2 diabetes: nature of early stage diabetes. Acta Diabetol 2013; 50:39-45. [PMID: 21861172 DOI: 10.1007/s00592-011-0319-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2010] [Accepted: 08/02/2011] [Indexed: 12/13/2022]
Abstract
At the time of diagnosis of type 2 diabetes (T2D), patients already have varying degrees of beta-cell dysfunction and insulin resistance and the defects continue to deteriorate despite treatment. We examined insulin secretion impairment and insulin resistance in overweight patients with T2D who had metformin failure, with elevated HbA1c at maximal metformin dose. Patients (N = 1,039) were examined at entry to the European Exenatide (EUREXA) clinical trial of add-on exenatide versus sulphonylurea. Mean (±SD) age was 57 ± 10 years, and BMI was 32.4 ± 4.1 kg/m(2). All patients underwent an oral glucose tolerance test; HOMA-IR, HOMA-B, ∆I(30)/∆G(30), disposition index and pro-insulin/insulin ratio were evaluated in relation to stratified HbA1c levels (≤7.3, >7.3-8.2, >8.2%) and duration of diabetes (<3, ≥3-<6, ≥6 years) using non-parametric analysis of variance. Patients overall had a wide range of impaired insulin secretion (HOMA-B: median 50.4, interquartile range 32.8-78.8) and insulin resistance (HOMA-IR: 4.8, 3.0-7.4). With increasing HbA1c levels, there was a statistically significant decrease in HOMA-B (P < 0.001), ∆I(30)/∆G(30) (P = 0.003) and disposition index (P < 0.001), and increase in pro-insulin/insulin (P < 0.001) and HOMA-IR (P < 0.001). With increasing duration since diabetes diagnosis, there was a significant decrease in HOMA-B (P < 0.001), but no significant trend in HOMA-IR, ∆I(30)/∆G(30), disposition index or pro-insulin/insulin. Metformin failure in these patients was associated with beta-cell dysfunction to a greater extent than insulin resistance. Loss of the first-phase insulin release, indicated by a low ∆I(30)/∆G(30), would indicate that this patient cohort requires add-on therapy that can maintain beta-cell function.
Collapse
Affiliation(s)
- Baptist Gallwitz
- Department of Medicine IV, Eberhard-Karls-University, Tübingen, Germany.
| | | | | | | | | |
Collapse
|
41
|
Chen H, Martin B, Cai H, Fiori JL, Egan JM, Siddiqui S, Maudsley S. Pancreas++: automated quantification of pancreatic islet cells in microscopy images. Front Physiol 2013; 3:482. [PMID: 23293605 PMCID: PMC3535421 DOI: 10.3389/fphys.2012.00482] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Accepted: 12/09/2012] [Indexed: 12/02/2022] Open
Abstract
The microscopic image analysis of pancreatic Islet of Langerhans morphology is crucial for the investigation of diabetes and metabolic diseases. Besides the general size of the islet, the percentage and relative position of glucagon-containing alpha-, and insulin-containing beta-cells is also important for pathophysiological analyses, especially in rodents. Hence, the ability to identify, quantify and spatially locate peripheral, and “involuted” alpha-cells in the islet core is an important analytical goal. There is a dearth of software available for the automated and sophisticated positional quantification of multiple cell types in the islet core. Manual analytical methods for these analyses, while relatively accurate, can suffer from a slow throughput rate as well as user-based biases. Here we describe a newly developed pancreatic islet analytical software program, Pancreas++, which facilitates the fully automated, non-biased, and highly reproducible investigation of islet area and alpha- and beta-cell quantity as well as position within the islet for either single or large batches of fluorescent images. We demonstrate the utility and accuracy of Pancreas++ by comparing its performance to other pancreatic islet size and cell type (alpha, beta) quantification methods. Our Pancreas++ analysis was significantly faster than other methods, while still retaining low error rates and a high degree of result correlation with the manually generated reference standard.
Collapse
Affiliation(s)
- Hongyu Chen
- Receptor Pharmacology Unit, Laboratory of Neuroscience, National Institute on Aging, National Institutes of Health Baltimore, MD, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Anichini R, Cosimi S, Di Carlo A, Orsini P, De Bellis A, Seghieri G, Franconi F, Baccetti F. Gender difference in response predictors after 1-year exenatide therapy twice daily in type 2 diabetic patients: a real world experience. Diabetes Metab Syndr Obes 2013; 6:123-9. [PMID: 23630427 PMCID: PMC3626369 DOI: 10.2147/dmso.s42729] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
PURPOSE To investigate whether gender affects therapeutic response by exenatide twice a day (BID) in type 2 diabetes by using a database concerning patients monitored by five outpatient clinics in Tuscany, Italy. PATIENTS AND METHODS We considered a cohort of 315 (154 male/161 female) patients experiencing therapeutic failure while on oral therapy (metformin, or combination therapy metformin + sulphonylureas), who were given exenatide (10 μg/BID) and who fully completed 4 months, 8 months, and 12 months of follow-ups. RESULTS Among patients stratified by gender and well matched for age, body mass index, and hemoglobin A1c (HbA1c), it was found that the length of disease was longer in females than in males (12 ± 8 years versus 10 ± 7 years; P = 0.037), and the ratio of patients on metformin to those on combination therapy was higher in men (P = 0.018). Target glycemic response (1-year HbA1c ≤ 7%) was achieved in a significantly higher proportion of males than females (38% versus 27%; χ(2) = 4.66; P = 0.03). Target weight loss expressed as 1-year weight percent fall from baseline ≥ 75th percentile (8.5%) was significantly higher in females at 8 and 12 months (P < 0.05; for both). One-year glycemic target response was inversely related to baseline HbA1c levels and diabetes duration among males, while metformin therapy (compared to oral combination therapy) was a significant predictor of better glycemic targets among females. Homeostasis model assessment-B, measured in 117 patients, predicted hypoglycemic response only in women (P = 0.009). Target 1-year weight loss was predicted by longer diabetes duration among males and by lower baseline HbA1c among females. Finally, no significant difference between genders was noted as to gastrointestinal side effects after exenatide therapy. CONCLUSION According to this "real world" experience, predictors of glycemic control and body weight loss after 12 months of exenatide BID therapy are different between genders in type 2 diabetes.
Collapse
Affiliation(s)
| | - Sabrina Cosimi
- Diabetes Unit, Hospital of Versilia, Camaiore (LU), Italy
| | | | | | | | - Giuseppe Seghieri
- Diabetes Unit, Spedali Riuniti, Pistoia, Italy
- Correspondence: Giuseppe Seghieri, Department of Internal Medicine, Spedali Riuniti, Viale Matteotti 9/D, 51100 Pistoia, Italy, Tel +39 338 694 1642, Fax +39 057 335 2005, Email
| | - Flavia Franconi
- Department of Biochemical Sciences, University of Sassari, Italy
| | | |
Collapse
|
43
|
Marchetti P, Bugliani M, Boggi U, Masini M, Marselli L. The pancreatic beta cells in human type 2 diabetes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 771:288-309. [PMID: 23393686 DOI: 10.1007/978-1-4614-5441-0_22] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Bell-cell (beta-cell) impairment is central to the development and progression of human diabetes, as a result of the combined effects of genetic and acquired factors. Reduced islet number and/or reduced beta cells amount in the pancreas of individuals with Type 2 diabetes have been consistently reported. This is mainly due to increased beta cell death, not adequately compensated for by regeneration. In addition, several quantitative and/or qualitative defects of insulin secretion have been observed in Type 2 diabetes, both in vivo and ex vivo with isolated islets. All this is associated with modifications of islet cell gene and protein expression. With the identification of several susceptible Type 2 diabetes loci, the role of genotype in affecting beta-cell function and survival has been addressed in a few studies and the relationships between genotype and beta-cell phenotype investigated. Among acquired factors, the importance of metabolic insults (in particular glucotoxicity and lipotoxicity) in the natural history of beta-cell damage has been widely underlined. Continuous improvements in our knowledge of the beta cells in human Type 2 diabetes will lead to more targeted and effective strategies for the prevention and treatment of the disease.
Collapse
Affiliation(s)
- Piero Marchetti
- Department of Endocrinology and Metabolism, University of Pisa, Pisa, Italy.
| | | | | | | | | |
Collapse
|
44
|
Ceccarelli E, Guarino EG, Merlotti D, Patti A, Gennari L, Nuti R, Dotta F. Beyond glycemic control in diabetes mellitus: effects of incretin-based therapies on bone metabolism. Front Endocrinol (Lausanne) 2013; 4:73. [PMID: 23785355 PMCID: PMC3684850 DOI: 10.3389/fendo.2013.00073] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 06/03/2013] [Indexed: 12/31/2022] Open
Abstract
Diabetes mellitus (DM) and osteoporosis (OP) are common disorders with a significant health burden, and an increase in fracture risk has been described both in type 1 (T1DM) and in type 2 (T2DM) diabetes. The pathogenic mechanisms of impaired skeletal strength in diabetes remain to be clarified in details and they are only in part reflected by a variation in bone mineral density. In T2DM, the occurrence of low bone turnover together with a decreased osteoblast activity and compromised bone quality has been shown. Of note, some antidiabetic drugs (e.g., thiazolidinediones, insulin) may deeply affect bone metabolism. In addition, the recently introduced class of incretin-based drugs (i.e., GLP-1 receptor agonists and DPP-4 inhibitors) is expected to exert potentially beneficial effects on bone health, possibly due to a bone anabolic activity of GLP-1, that can be either direct or indirect through the involvement of thyroid C cells. Here we will review the established as well as the putative effects of incretin hormones and of incretin-based drugs on bone metabolism, both in preclinical models and in man, taking into account that such therapeutic strategy may be effective not only to achieve a good glycemic control, but also to improve bone health in diabetic patients.
Collapse
Affiliation(s)
- Elena Ceccarelli
- Diabetes Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Elisa G. Guarino
- Diabetes Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Daniela Merlotti
- Internal Medicine Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Aurora Patti
- Diabetes Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
- Internal Medicine Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Luigi Gennari
- Internal Medicine Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Ranuccio Nuti
- Internal Medicine Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Francesco Dotta
- Diabetes Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
- Fondazione Umberto Di Mario ONLUS, Siena, Italy
- *Correspondence: Francesco Dotta, U.O.C. Diabetologia, University of Siena, Policlinico Le Scotte, Viale Bracci 18, Siena 53100, Italy e-mail:
| |
Collapse
|
45
|
Masini M, Marselli L, Bugliani M, Martino L, Masiello P, Marchetti P, De Tata V. Ultrastructural morphometric analysis of insulin secretory granules in human type 2 diabetes. Acta Diabetol 2012. [PMID: 23184237 DOI: 10.1007/s00592-012-0446-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
We performed an ultrastructural morphometric analysis of insulin secretory granules in pancreatic beta cells from control and type 2 diabetic multiorgan donors. The volume density of insulin granules significantly (p < 0.05) reduced in beta cells from type 2 diabetic patients with respect to non-diabetic subjects, and this reduction was mainly attributable to a decrease in mature granules. On the contrary, no significant difference was observed in the volume density of docked granules between controls and type 2 diabetic patients. In addition, there was a significant positive correlation between the density volume of total insulin granules and stimulated insulin secretion in non-diabetic islets. In conclusion, we detected significant changes in the intracellular distribution of insulin secretory granules within the beta cell that might be related with the alterations in insulin secretion observed in type 2 diabetes patients.
Collapse
Affiliation(s)
- Matilde Masini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, Scuola Medica, 56126 Pisa, Italy
| | | | | | | | | | | | | |
Collapse
|
46
|
Yeo RWY, Yang K, Li G, Lim SK. High glucose predisposes gene expression and ERK phosphorylation to apoptosis and impaired glucose-stimulated insulin secretion via the cytoskeleton. PLoS One 2012; 7:e44988. [PMID: 23024780 PMCID: PMC3443235 DOI: 10.1371/journal.pone.0044988] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 08/11/2012] [Indexed: 01/09/2023] Open
Abstract
Chronic high glucose (HG) inflicts glucotoxicity on vulnerable cell types such as pancreatic β cells and contributes to insulin resistance and impaired insulin secretion in diabetic patients. To identify HG-induced cellular aberrations that are candidate mediators of glucotoxicity in pancreatic β cells, we analyzed gene expression in ERoSHK6, a mouse insulin-secreting cell line after chronic HG exposure (six-day exposure to 33.3 mM glucose). Chronic HG exposure which reduced glucose-stimulated insulin secretion (GSIS) increased transcript levels of 185 genes that clustered primarily in 5 processes namely cellular growth and proliferation; cell death; cellular assembly and organization; cell morphology; and cell-to-cell signaling and interaction. The former two were validated by increased apoptosis of ERoSHK6 cells after chronic HG exposure and reaffirmed the vulnerability of β cells to glucotoxicity. The three remaining processes were partially substantiated by changes in cellular morphology and structure, and instigated an investigation of the cytoskeleton and cell-cell adhesion. These studies revealed a depolymerized actin cytoskeleton that lacked actin stress fibers anchored at vinculin-containing focal adhesion sites as well as loss of E-cadherin-mediated cell-cell adherence after exposure to chronic HG, and were concomitant with constitutive ERK1/2 phosphorylation that was refractory to serum and glucose deprivation. Although inhibition of ERK phosphorylation by PD98059 promoted actin polymerization, it increased apoptosis and GSIS impairment. These findings suggest that ERK phosphorylation is a proximate regulator of cellular processes targeted by chronic HG-induced gene expression and that dynamic actin polymerization and depolymerization is important in β cell survival and function. Therefore, chronic HG alters gene expression and signal transduction to predispose the cytoskeleton towards apoptosis and GSIS impairment.
Collapse
Affiliation(s)
- Ronne Wee Yeh Yeo
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, Singapore
| | | | - GuoDong Li
- Department of Clinical Research, Singapore General Hospital, Singapore, Singapore
| | - Sai Kiang Lim
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- * E-mail:
| |
Collapse
|
47
|
Abstract
Polygenic type 2 diabetes mellitus (T2DM) is a multi-factorial disease due to the interplay between genes and the environment. Over the years, several genes/loci have been associated with this type of diabetes, with the majority of them being related to β cell dysfunction. In this review, the available information on how polymorphisms in T2DM-associated genes/loci do directly affect the properties of human islet cells are presented and discussed, including some clinical implications and the role of epigenetic mechanisms.
Collapse
Affiliation(s)
- Piero Marchetti
- Department of Endocrinology and Metabolism, University of Pisa, Pisa, Italy.
| | | | | | | | | |
Collapse
|
48
|
Chung JO, Cho DH, Chung DJ, Chung MY. Associations between hemoglobin concentrations and the clinical characteristics of patients with type 2 diabetes. Korean J Intern Med 2012; 27:285-92. [PMID: 23019393 PMCID: PMC3443721 DOI: 10.3904/kjim.2012.27.3.285] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Revised: 02/03/2012] [Accepted: 04/23/2012] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND/AIMS Many studies have demonstrated an association between hemoglobin levels and cardiovascular disease in diabetic patients. The aim of this study was to determine whether there is an association between hemoglobin concentrations and various clinical parameters, including metabolic factors, plasma C-peptide response after a meal tolerance test, and microvascular complications, in Korean patients with type 2 diabetes. METHODS In total, 337 male patients with type 2 diabetes were recruited. All subjects were subjected to a meal tolerance test and underwent assessment of hemoglobin levels, fasting and postprandial β-cell responsiveness, and microvascular complications. RESULTS Patients with lower hemoglobin concentrations had a longer duration of diabetes, a lower body mass index, and lower concentrations of total cholesterol, triglycerides, and low-density lipoprotein cholesterol. They also had lower levels of postprandial C-peptide, Δ C-peptide, and postprandial β-cell responsiveness. They had a higher prevalence of retinopathy and nephropathy. In multivariate analyses, there was a significant association between nephropathy and hemoglobin concentration. Also, hemoglobin concentrations were independently associated with Δ C-peptide levels and postprandial β-cell responsiveness. CONCLUSIONS Hemoglobin concentrations are associated with postprandial C-peptide responses and diabetic nephropathy in patients with type 2 diabetes.
Collapse
Affiliation(s)
- Jin Ook Chung
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Dong Hyeok Cho
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Dong Jin Chung
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Min Young Chung
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| |
Collapse
|
49
|
Carlsson LMS, Peltonen M, Ahlin S, Anveden Å, Bouchard C, Carlsson B, Jacobson P, Lönroth H, Maglio C, Näslund I, Pirazzi C, Romeo S, Sjöholm K, Sjöström E, Wedel H, Svensson PA, Sjöström L. Bariatric surgery and prevention of type 2 diabetes in Swedish obese subjects. N Engl J Med 2012; 367:695-704. [PMID: 22913680 DOI: 10.1056/nejmoa1112082] [Citation(s) in RCA: 514] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Weight loss protects against type 2 diabetes but is hard to maintain with behavioral modification alone. In an analysis of data from a nonrandomized, prospective, controlled study, we examined the effects of bariatric surgery on the prevention of type 2 diabetes. METHODS In this analysis, we included 1658 patients who underwent bariatric surgery and 1771 obese matched controls (with matching performed on a group, rather than individual, level). None of the participants had diabetes at baseline. Patients in the bariatric-surgery cohort underwent banding (19%), vertical banded gastroplasty (69%), or gastric bypass (12%); nonrandomized, matched, prospective controls received usual care. Participants were 37 to 60 years of age, and the body-mass index (BMI; the weight in kilograms divided by the square of the height in meters) was 34 or more in men and 38 or more in women. This analysis focused on the rate of incident type 2 diabetes, which was a prespecified secondary end point in the main study. At the time of this analysis (January 1, 2012), participants had been followed for up to 15 years. Despite matching, some baseline characteristics differed significantly between the groups; the baseline body weight was higher and risk factors were more pronounced in the bariatric-surgery group than in the control group. At 15 years, 36.2% of the original participants had dropped out of the study, and 30.9% had not yet reached the time for their 15-year follow-up examination. RESULTS During the follow-up period, type 2 diabetes developed in 392 participants in the control group and in 110 in the bariatric-surgery group, corresponding to incidence rates of 28.4 cases per 1000 person-years and 6.8 cases per 1000 person-years, respectively (adjusted hazard ratio with bariatric surgery, 0.17; 95% confidence interval, 0.13 to 0.21; P<0.001). The effect of bariatric surgery was influenced by the presence or absence of impaired fasting glucose (P=0.002 for the interaction) but not by BMI (P=0.54). Sensitivity analyses, including end-point imputations, did not change the overall conclusions. The postoperative mortality was 0.2%, and 2.8% of patients who underwent bariatric surgery required reoperation within 90 days owing to complications. CONCLUSIONS Bariatric surgery appears to be markedly more efficient than usual care in the prevention of type 2 diabetes in obese persons. (Funded by the Swedish Research Council and others; ClinicalTrials.gov number, NCT01479452.).
Collapse
Affiliation(s)
- Lena M S Carlsson
- Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Erejuwa OO, Sulaiman SA, Wahab MSA. Honey--a novel antidiabetic agent. Int J Biol Sci 2012; 8:913-34. [PMID: 22811614 PMCID: PMC3399220 DOI: 10.7150/ijbs.3697] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2011] [Accepted: 01/24/2012] [Indexed: 12/26/2022] Open
Abstract
Diabetes mellitus remains a burden worldwide in spite of the availability of numerous antidiabetic drugs. Honey is a natural substance produced by bees from nectar. Several evidence-based health benefits have been ascribed to honey in the recent years. In this review article, we highlight findings which demonstrate the beneficial or potential effects of honey in the gastrointestinal tract (GIT), on the gut microbiota, in the liver, in the pancreas and how these effects could improve glycemic control and metabolic derangements. In healthy subjects or patients with impaired glucose tolerance or diabetes mellitus, various studies revealed that honey reduced blood glucose or was more tolerable than most common sugars or sweeteners. Pre-clinical studies provided more convincing evidence in support of honey as a potential antidiabetic agent than clinical studies did. The not-too-impressive clinical data could mainly be attributed to poor study designs or due to the fact that the clinical studies were preliminary. Based on the key constituents of honey, the possible mechanisms of action of antidiabetic effect of honey are proposed. The paper also highlights the potential impacts and future perspectives on the use of honey as an antidiabetic agent. It makes recommendations for further clinical studies on the potential antidiabetic effect of honey. This review provides insight on the potential use of honey, especially as a complementary agent, in the management of diabetes mellitus. Hence, it is very important to have well-designed, randomized controlled clinical trials that investigate the reproducibility (or otherwise) of these experimental data in diabetic human subjects.
Collapse
Affiliation(s)
- Omotayo O Erejuwa
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia.
| | | | | |
Collapse
|