1
|
Damiani V, Giuseppe GD, Gliozzo G, Ciccarelli G, Pizzinato E, Pizzo FD, Fruci D, Brunetti M, Soldovieri L, Quero G, Mari A, Alfieri S, Pontecorvi A, Giaccari A, Laurenzi VD, Mezza T. Altered BAG3-insulin colocalization is associated with impaired first phase insulin secretion in humans. Diabetes Res Clin Pract 2025:112232. [PMID: 40339705 DOI: 10.1016/j.diabres.2025.112232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 03/28/2025] [Accepted: 05/03/2025] [Indexed: 05/10/2025]
Abstract
AIMS Alterations in first-phase insulin secretion are pivotal in the early development of T2DM. BAG3 has been implicated in regulating insulin secretion in murine models, but its role in humans remains unexplored. This study investigates BAG3 expression in human pancreatic islets and its relationship with β-cell functionality. METHODS Pancreatic tissue samples were obtained from 12 patients with no previous T2DM diagnosis enrolled for partial pancreatectomy. Patients underwent deep metabolic evaluation, including OGTT, hyperglycemic clamp and euglycemic hyperinsulinemic clamp. Immunofluorescence and confocal microscopy were used to assess BAG3-insulin colocalization and further correlated with metabolic findings, categorizing subjects into LOW and HIGH BAG3 groups. RESULTS Patients with HIGH BAG3 expression exhibited significantly impaired first-phase insulin secretion, evidenced by reduced rate sensitivity during OGTT and higher plasma glucose levels at 30 and 60 min post-glucose challenge. Islets from HIGH BAG3 patients showed increased size but no differences in insulin/glucagon ratios or insulin sensitivity, suggesting a specific disruption in the insulin secretory machinery rather than β-cell mass or insulin resistance. CONCLUSIONS BAG3 appears associated to first-phase insulin secretion in humans by influencing insulin granule exocytosis. Targeting BAG3 could represent a novel therapeutic approach to prevent or delay β-cell dysfunction and the onset of T2DM.
Collapse
Affiliation(s)
- Verena Damiani
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy; Department of Innovative Technologies in Medicine and Dentistry, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy; Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Gianfranco Di Giuseppe
- Endocrinology and Diabetology Unit, University Hospital Agostino Gemelli, Rome, Italy; Department of Translational Medicine and Surgery, Catholic University of Sacred Heart, Rome, Italy
| | - Giulia Gliozzo
- Department of Translational Medicine and Surgery, Catholic University of Sacred Heart, Rome, Italy
| | - Gea Ciccarelli
- Endocrinology and Diabetology Unit, University Hospital Agostino Gemelli, Rome, Italy; Department of Translational Medicine and Surgery, Catholic University of Sacred Heart, Rome, Italy
| | - Erika Pizzinato
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy; Department of Innovative Technologies in Medicine and Dentistry, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy; Telematic University of "Leonardo Da Vinci", Torrevecchia Teatina, Chieti Telematic University of "Leonardo Da Vinci", Torrevecchia Teatina, Chieti, Italy
| | - Francesco Del Pizzo
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy; Department of Innovative Technologies in Medicine and Dentistry, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Doriana Fruci
- Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Michela Brunetti
- Endocrinology and Diabetology Unit, University Hospital Agostino Gemelli, Rome, Italy; Department of Translational Medicine and Surgery, Catholic University of Sacred Heart, Rome, Italy
| | - Laura Soldovieri
- Endocrinology and Diabetology Unit, University Hospital Agostino Gemelli, Rome, Italy; Department of Translational Medicine and Surgery, Catholic University of Sacred Heart, Rome, Italy
| | - Giuseppe Quero
- Department of Translational Medicine and Surgery, Catholic University of Sacred Heart, Rome, Italy; Digestive Surgery Unit, University Hospital Agostino Gemelli, Rome, Italy
| | - Andrea Mari
- Institute of Neuroscience, National Research Council, Padua, Italy
| | - Sergio Alfieri
- Department of Translational Medicine and Surgery, Catholic University of Sacred Heart, Rome, Italy; Digestive Surgery Unit, University Hospital Agostino Gemelli, Rome, Italy
| | - Alfredo Pontecorvi
- Endocrinology and Diabetology Unit, University Hospital Agostino Gemelli, Rome, Italy; Department of Translational Medicine and Surgery, Catholic University of Sacred Heart, Rome, Italy
| | - Andrea Giaccari
- Endocrinology and Diabetology Unit, University Hospital Agostino Gemelli, Rome, Italy; Department of Translational Medicine and Surgery, Catholic University of Sacred Heart, Rome, Italy.
| | - Vincenzo De Laurenzi
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy; Department of Innovative Technologies in Medicine and Dentistry, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy.
| | - Teresa Mezza
- Endocrinology and Diabetology Unit, University Hospital Agostino Gemelli, Rome, Italy; Department of Translational Medicine and Surgery, Catholic University of Sacred Heart, Rome, Italy
| |
Collapse
|
2
|
Jo S, Jing G, Chen J, Xu G, Shalev A. Oral TIX100 protects against obesity-associated glucose intolerance and diet-induced adiposity. Diabetes Obes Metab 2025; 27:2223-2231. [PMID: 39895486 PMCID: PMC11885086 DOI: 10.1111/dom.16223] [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: 11/03/2024] [Revised: 01/11/2025] [Accepted: 01/21/2025] [Indexed: 02/04/2025]
Abstract
AIMS Glucagon-like peptide 1 receptor agonists and dual agonists have changed the treatment landscape of obesity and type 2 diabetes (T2D), but significant limitations have emerged due to their gastrointestinal side effects, loss of lean mass, and necessity for ongoing subcutaneous injections. Our objective was, therefore, to test a novel small molecule as a different and potentially better tolerated oral medications to improve obesity-associated impairment in glucose homeostasis. MATERIALS AND METHODS High-fat diet (HFD)-fed mice or severely obese, leptin-deficient ob/ob mice were randomly assigned to serve as controls or receive oral TIX100, a novel thioredoxin-interacting protein (TXNIP) inhibitor just approved by the FDA as an investigational new drug for type 1 diabetes (T1D). The TIX100 effects on glucose intolerance and weight control were then assessed. RESULTS TIX100 protected against HFD-induced glucose intolerance, hyperinsulinemia, and hyperglucagonemia. TIX100 also reduced diet-induced adiposity resulting in 15% lower weight in treated mice as compared with controls on HFD (p <0.05), while preserving lean mass. Even though the TIX100 weight effects were lost in ob/ob mice, TIX100 improved glucose control leading to a dramatic 2.3% reduction in HbA1C (p <0.05), independent of any weight loss. This is consistent with the beneficial effects of TIX100 in non-obese diabetes models and its protection against elevated TXNIP and islet cell stress common to all diabetes types. CONCLUSIONS Thus, TIX100 may provide a novel, oral therapy for T2D that targets underlying disease pathology including islet cell dysfunction and hyperglucagonemia and promotes metabolic health and weight control without aggressive weight loss.
Collapse
Affiliation(s)
- SeongHo Jo
- Comprehensive Diabetes Center, Department of Medicine, Division of Endocrinology, Diabetes, and MetabolismUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Gu Jing
- Comprehensive Diabetes Center, Department of Medicine, Division of Endocrinology, Diabetes, and MetabolismUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Junqin Chen
- Comprehensive Diabetes Center, Department of Medicine, Division of Endocrinology, Diabetes, and MetabolismUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Guanlan Xu
- Comprehensive Diabetes Center, Department of Medicine, Division of Endocrinology, Diabetes, and MetabolismUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Anath Shalev
- Comprehensive Diabetes Center, Department of Medicine, Division of Endocrinology, Diabetes, and MetabolismUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| |
Collapse
|
3
|
Sudhayakumar A, Arjunan D, Bhansali S, Bhujade H, Bhadada SK, Malhotra S, Walia R. Realisation of Remission of Diabetes Using Pharmacotherapy (DiaRem-1). Indian J Endocrinol Metab 2025; 29:217-223. [PMID: 40416453 PMCID: PMC12101762 DOI: 10.4103/ijem.ijem_356_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 02/22/2025] [Accepted: 02/27/2025] [Indexed: 05/27/2025] Open
Abstract
Introduction Type 2 diabetes mellitus has traditionally been considered a lifelong disease, with treatment focused on glycaemic control and complication prevention. Emerging evidence suggests that targeting glucolipotoxicity and insulin resistance may restore beta-cell function, potentially leading to remission. This study aimed to evaluate the feasibility of achieving diabetes remission through pharmacotherapy by promoting weight loss and maintaining strict glycaemic control over a 3-month period. Methods This is a single-centre, open-label, randomised controlled trial conducted at a tertiary care center in India, with adult subjects with type 2 diabetes mellitus <5 years duration and HbA1c <8.5% randomised to an intervention arm using liraglutide, dapagliflozin and metformin, and a control arm using vildagliptin, glimepiride and metformin. The subjects were treated for 3 months followed by an off-treatment period for 3 months to assess for remission of diabetes. Results Two hundred thirty-six subjects were assessed for eligibility and 29 subjects underwent randomisation. Fourteen subjects were randomised to the intervention arm and 15 subjects were randomised to the control arm. Twenty-three of the 29 recruited subjects completed the trial. At the end of the off-treatment period, nine patients (31.04%) were in remission, with 4 out of 14 patients (28.57%) in the intervention arm and 5 out of 15 patients (33.33%) in the control arm maintaining HbA1c less than 6.5% without any treatment. No baseline clinical or biochemical parameters were found to be reliable predictors of remission. Conclusion This trial provides evidence that pharmacotherapy targeting tight glycaemic control on an outpatient basis is effective in achieving diabetes remission.
Collapse
Affiliation(s)
| | | | | | - Harish Bhujade
- Department of Radiodiagnosis and Imaging, PGIMER, Chandigarh, India
| | | | | | - Rama Walia
- Department of Endocrinology, PGIMER, Chandigarh, India
| |
Collapse
|
4
|
Korvyakova Y, Azarova I, Klyosova E, Postnikova M, Makarenko V, Bushueva O, Solodilova M, Polonikov A. The link between the ANPEP gene and type 2 diabetes mellitus may be mediated by the disruption of glutathione metabolism and redox homeostasis. Gene 2025; 935:149050. [PMID: 39489227 DOI: 10.1016/j.gene.2024.149050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 10/02/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
Aminopeptidase N (ANPEP), a membrane-associated ectoenzyme, has been identified as a susceptibility gene for type 2 diabetes (T2D) by genome-wide association and transcriptome studies; however, the mechanisms by which this gene contributes to disease pathogenesis remain unclear. The aim of this study was to determine the comprehensive contribution of ANPEP polymorphisms to T2D risk and annotate the underlying mechanisms. A total of 3206 unrelated individuals including 1579 T2D patients and 1627 controls were recruited for the study. Twenty-three common functional single nucleotide polymorphisms (SNP) of ANPEP were genotyped by the MassArray-4 system. Six polymorphisms, rs11073891, rs12898828, rs12148357, rs9920421, rs7111, and rs25653, were found to be associated with type 2 diabetes (Pperm ≤ 0.05). Common haplotype rs9920421G-rs4932143G-rs7111T was strongly associated with increased risk of T2D (Pperm = 5.9 × 10-12), whereas two rare haplotypes such as rs9920421G-rs4932143C-rs7111T (Pperm = 6.5 × 10-40) and rs12442778A-rs12898828A-rs6496608T-rs11073891C (Pperm = 1.0 × 10-7) possessed strong protection against disease. We identified 38 and 109 diplotypes associated with T2D risk in males and females, respectively (FDR ≤ 0.05). ANPEP polymorphisms showed associations with plasma levels of fasting blood glucose, aspartate aminotransferase, total protein and glutathione (P < 0.05), and several haplotypes were strongly associated with the levels of reactive oxygen species and uric acid (P < 0.0001). A deep literature analysis has facilitated the formulation of a hypothesis proposing that increased plasma levels of ANPEP as well as liver enzymes such as aspartate aminotransferase, alanine aminotransferase and gammaglutamyltransferase serve as an adaptive response directed towards the restoration of glutathione deficiency in diabetics by stimulating the production of amino acid precursors for glutathione biosynthesis.
Collapse
Affiliation(s)
- Yaroslava Korvyakova
- Laboratory of Biochemical Genetics and Metabolomics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russian Federation; Research Centre for Medical Genetics, 1 Moskvorechie St., Moscow 115522, Russian Federation.
| | - Iuliia Azarova
- Laboratory of Biochemical Genetics and Metabolomics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russian Federation; Department of Biological Chemistry, Kursk State Medical University, 3 Karl Marx Street, Kursk 305041, Russian Federation.
| | - Elena Klyosova
- Laboratory of Biochemical Genetics and Metabolomics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russian Federation; Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx Street, Kursk 305041, Russian Federation.
| | - Maria Postnikova
- Laboratory of Biochemical Genetics and Metabolomics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russian Federation.
| | - Victor Makarenko
- Laboratory of Biochemical Genetics and Metabolomics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russian Federation
| | - Olga Bushueva
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx Street, Kursk 305041, Russian Federation; Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russian Federation.
| | - Maria Solodilova
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx Street, Kursk 305041, Russian Federation.
| | - Alexey Polonikov
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx Street, Kursk 305041, Russian Federation; Laboratory of Statistical Genetics and Bioinformatics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russian Federation.
| |
Collapse
|
5
|
Grenko CM, Taylor HJ, Bonnycastle LL, Xue D, Lee BN, Weiss Z, Yan T, Swift AJ, Mansell EC, Lee A, Robertson CC, Narisu N, Erdos MR, Chen S, Collins FS, Taylor DL. Single-cell transcriptomic profiling of human pancreatic islets reveals genes responsive to glucose exposure over 24 h. Diabetologia 2024; 67:2246-2259. [PMID: 38967666 PMCID: PMC11447040 DOI: 10.1007/s00125-024-06214-4] [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: 03/04/2024] [Accepted: 05/08/2024] [Indexed: 07/06/2024]
Abstract
AIMS/HYPOTHESIS Disruption of pancreatic islet function and glucose homeostasis can lead to the development of sustained hyperglycaemia, beta cell glucotoxicity and subsequently type 2 diabetes. In this study, we explored the effects of in vitro hyperglycaemic conditions on human pancreatic islet gene expression across 24 h in six pancreatic cell types: alpha; beta; gamma; delta; ductal; and acinar. We hypothesised that genes associated with hyperglycaemic conditions may be relevant to the onset and progression of diabetes. METHODS We exposed human pancreatic islets from two donors to low (2.8 mmol/l) and high (15.0 mmol/l) glucose concentrations over 24 h in vitro. To assess the transcriptome, we performed single-cell RNA-seq (scRNA-seq) at seven time points. We modelled time as both a discrete and continuous variable to determine momentary and longitudinal changes in transcription associated with islet time in culture or glucose exposure. Additionally, we integrated genomic features and genetic summary statistics to nominate candidate effector genes. For three of these genes, we functionally characterised the effect on insulin production and secretion using CRISPR interference to knock down gene expression in EndoC-βH1 cells, followed by a glucose-stimulated insulin secretion assay. RESULTS In the discrete time models, we identified 1344 genes associated with time and 668 genes associated with glucose exposure across all cell types and time points. In the continuous time models, we identified 1311 genes associated with time, 345 genes associated with glucose exposure and 418 genes associated with interaction effects between time and glucose across all cell types. By integrating these expression profiles with summary statistics from genetic association studies, we identified 2449 candidate effector genes for type 2 diabetes, HbA1c, random blood glucose and fasting blood glucose. Of these candidate effector genes, we showed that three (ERO1B, HNRNPA2B1 and RHOBTB3) exhibited an effect on glucose-stimulated insulin production and secretion in EndoC-βH1 cells. CONCLUSIONS/INTERPRETATION The findings of our study provide an in-depth characterisation of the 24 h transcriptomic response of human pancreatic islets to glucose exposure at a single-cell resolution. By integrating differentially expressed genes with genetic signals for type 2 diabetes and glucose-related traits, we provide insights into the molecular mechanisms underlying glucose homeostasis. Finally, we provide functional evidence to support the role of three candidate effector genes in insulin secretion and production. DATA AVAILABILITY The scRNA-seq data from the 24 h glucose exposure experiment performed in this study are available in the database of Genotypes and Phenotypes (dbGap; https://www.ncbi.nlm.nih.gov/gap/ ) with accession no. phs001188.v3.p1. Study metadata and summary statistics for the differential expression, gene set enrichment and candidate effector gene prediction analyses are available in the Zenodo data repository ( https://zenodo.org/ ) under accession number 11123248. The code used in this study is publicly available at https://github.com/CollinsLabBioComp/publication-islet_glucose_timecourse .
Collapse
Affiliation(s)
- Caleb M Grenko
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
- Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, USA
| | - Henry J Taylor
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA.
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- Heart and Lung Research Institute, University of Cambridge, Cambridge, UK.
| | - Lori L Bonnycastle
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Dongxiang Xue
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
- Center for Genomic Health, Weill Cornell Medicine, New York, NY, USA
| | - Brian N Lee
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Zoe Weiss
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tingfen Yan
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Amy J Swift
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Erin C Mansell
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Angela Lee
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Catherine C Robertson
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Narisu Narisu
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Michael R Erdos
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Shuibing Chen
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
- Center for Genomic Health, Weill Cornell Medicine, New York, NY, USA
| | - Francis S Collins
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA.
| | - D Leland Taylor
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
6
|
Chan JCN, Mbanya JC, Chantelot J, Shestakova M, Ramachandran A, Ilkova H, Deplante L, Rollot M, Melas‐Melt L, Gagliardino JJ, Aschner P. Patient-reported outcomes and treatment adherence in type 2 diabetes using natural language processing: Wave 8 of the Observational International Diabetes Management Practices Study. J Diabetes Investig 2024; 15:1306-1316. [PMID: 38840439 PMCID: PMC11363103 DOI: 10.1111/jdi.14228] [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: 11/07/2023] [Revised: 04/17/2024] [Accepted: 04/20/2024] [Indexed: 06/07/2024] Open
Abstract
AIMS/INTRODUCTION We analyzed patient-reported outcomes of people with type 2 diabetes to better understand perceptions and experiences contributing to treatment adherence. MATERIALS AND METHODS In the ongoing International Diabetes Management Practices Study, we collected patient-reported outcomes data from structured questionnaires (chronic treatment acceptance questionnaire and Diabetes Self-Management Questionnaire) and free-text answers to open-ended questions to assess perceptions of treatment value and side-effects, as well as barriers to, and enablers for, adherence and self-management. Free-text answers were analyzed by natural language processing. RESULTS In 2018-2020, we recruited 2,475 patients with type 2 diabetes (43.3% insulin-treated, glycated hemoglobin (HbA1c) 8.0 ± 1.8%; 30.9% with HbA1c <7%) from 13 countries across Africa, the Middle East, Europe, Latin America and Asia. Mean ± standard deviation scores of chronic treatment acceptance questionnaire (acceptance of medication, rated out of 100) and Diabetes Self-Management Questionnaire (self-management, rated out of 10) were 87.8 ± 24.5 and 3.3 ± 0.9, respectively. Based on free-text analysis and coded responses, one in three patients reported treatment non-adherence. Overall, although most patients accepted treatment values and side-effects, self-management was suboptimal. Treatment duration, regimen complexity and disruption of daily routines were major barriers to adherence, whereas habit formation was a key enabler. Treatment-adherent patients were older (60 ± 11.6 vs 55 ± 11.7 years, P < 0.001), and more likely to have longer disease duration (12 ± 8.6 vs 10 ± 7.7 years, P < 0.001), exposure to diabetes education (73.1% vs 67.8%, P < 0.05), lower HbA1c (7.9 ± 1.8% vs 8.3 ± 1.9%, P < 0.001) and attainment of HbA1c <7% (29.7% vs 23.3%, P < 0.01). CONCLUSIONS Patient perceptions/experiences influence treatment adherence and self-management. Patient-centered education and support programs that consider patient-reported outcomes aimed at promoting empowerment and developing new routines might improve glycemic control.
Collapse
Affiliation(s)
- Juliana CN Chan
- Department of Medicine and TherapeuticsHong Kong Institute of Diabetes and Obesity and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales HospitalHong Kong SARChina
| | - Jean Claude Mbanya
- Doctoral School of Life Sciences, Health and Environment, and Department of Medicine and Specialties, Faculty of Medicine and Biomedical SciencesUniversity of Yaoundé IYaoundéCameroon
| | | | | | - Ambady Ramachandran
- India Diabetes Research Foundation, Dr. A. Ramachandran's Diabetes HospitalsChennaiIndia
| | | | - Lucille Deplante
- Quinten HealthParisFrance
- Present address:
49 Avenue Jean Jaurès38500VoironFrance
| | | | | | | | - Pablo Aschner
- Javeriana University School of Medicine and San Ignacio University HospitalBogotáColombia
| |
Collapse
|
7
|
Mei J, Li Y, Niu L, Liang R, Tang M, Cai Q, Xu J, Zhang D, Yin X, Liu X, Shen Y, Liu J, Xu M, Xia P, Ling J, Wu Y, Liang J, Zhang J, Yu P. SGLT2 inhibitors: a novel therapy for cognitive impairment via multifaceted effects on the nervous system. Transl Neurodegener 2024; 13:41. [PMID: 39123214 PMCID: PMC11312905 DOI: 10.1186/s40035-024-00431-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 07/11/2024] [Indexed: 08/12/2024] Open
Abstract
The rising prevalence of diabetes mellitus has casted a spotlight on one of its significant sequelae: cognitive impairment. Sodium-glucose cotransporter-2 (SGLT2) inhibitors, originally developed for diabetes management, are increasingly studied for their cognitive benefits. These benefits may include reduction of oxidative stress and neuroinflammation, decrease of amyloid burdens, enhancement of neuronal plasticity, and improved cerebral glucose utilization. The multifaceted effects and the relatively favorable side-effect profile of SGLT2 inhibitors render them a promising therapeutic candidate for cognitive disorders. Nonetheless, the application of SGLT2 inhibitors for cognitive impairment is not without its limitations, necessitating more comprehensive research to fully determine their therapeutic potential for cognitive treatment. In this review, we discuss the role of SGLT2 in neural function, elucidate the diabetes-cognition nexus, and synthesize current knowledge on the cognitive effects of SGLT2 inhibitors based on animal studies and clinical evidence. Research gaps are proposed to spur further investigation.
Collapse
Affiliation(s)
- Jiaqi Mei
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Huan Kui College of Nanchang University, Nanchang, China
| | - Yi Li
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Huan Kui College of Nanchang University, Nanchang, China
| | - Liyan Niu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Huan Kui College of Nanchang University, Nanchang, China
| | - Ruikai Liang
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Mingyue Tang
- Queen Mary College of Nanchang University, Nanchang, China
| | - Qi Cai
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jingdong Xu
- Queen Mary College of Nanchang University, Nanchang, China
| | - Deju Zhang
- Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Xiaoping Yin
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, China
| | - Xiao Liu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yunfeng Shen
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jianping Liu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Minxuan Xu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Panpan Xia
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jitao Ling
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yuting Wu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jianqi Liang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.
| | - Peng Yu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China.
| |
Collapse
|
8
|
Baghestani S, Haldin C, Kosijer P, Alam CM, Toivola DM. β-Cell keratin 8 maintains islet mechanical integrity, mitochondrial ultrastructure, and β-cell glucose transporter 2 plasma membrane targeting. Am J Physiol Cell Physiol 2024; 327:C462-C476. [PMID: 38912736 DOI: 10.1152/ajpcell.00123.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/31/2024] [Accepted: 06/12/2024] [Indexed: 06/25/2024]
Abstract
Islet β-cell dysfunction is an underlying factor for type I diabetes (T1D) development. Insulin sensing and secretion are tightly regulated in β-cells at multiple subcellular levels. The epithelial intermediate filament (IF) protein keratin (K) 8 is the main β-cell keratin, constituting the filament network with K18. To identify the cell-autonomous functions of K8 in β-cells, mice with targeted deletion of β-cell K8 (K8flox/flox; Ins-Cre) were analyzed for islet morphology, ultrastructure, and integrity, as well as blood glucose regulation and streptozotocin (STZ)-induced diabetes development. Glucose transporter 2 (GLUT2) localization was studied in β-cells in vivo and in MIN6 cells with intact or disrupted K8/K18 filaments. Loss of β-cell K8 leads to a major reduction in K18. Islets without β-cell K8 are more fragile, and these β-cells display disjointed plasma membrane organization with less membranous E-cadherin and smaller mitochondria with diffuse cristae. Lack of β-cell K8 also leads to a reduced glucose-stimulated insulin secretion (GSIS) response in vivo, despite undisturbed systemic blood glucose regulation. K8flox/flox, Ins-Cre mice have a decreased sensitivity to STZ compared with K8 wild-type mice, which is in line with decreased membranous GLUT2 expression observed in vivo, as GLUT2 is required for STZ uptake in β-cells. In vitro, MIN6 cell plasma membrane GLUT2 is rescued in cells overexpressing K8/K18 filaments but mistargeted in cells with disrupted K8/K18 filaments. β-Cell K8 is required for islet and β-cell structural integrity, normal mitochondrial morphology, and GLUT2 plasma membrane targeting, and has implications on STZ sensitivity as well as systemic insulin responses.NEW & NOTEWORTHY Keratin 8 is the main cytoskeletal protein in the cytoplasmic intermediate filament network in β-cells. Here for the first time, we assessed the β-cell autonomous mechanical and nonmechanical roles of keratin 8 in β-cell function. We demonstrated the importance of keratin 8 in islet and β-cell structural integrity, maintaining mitochondrial morphology and GLUT2 plasma membrane targeting.
Collapse
Affiliation(s)
- Sarah Baghestani
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship, Åbo Akademi University, Turku, Finland
| | - Caroline Haldin
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship, Åbo Akademi University, Turku, Finland
| | - Petar Kosijer
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship, Åbo Akademi University, Turku, Finland
| | - Catharina M Alam
- School of Applied Sciences, Edinburgh Napier University, Edinburg, United Kingdom
| | - Diana M Toivola
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship, Åbo Akademi University, Turku, Finland
- Turku Center for Disease Modeling, University of Turku, Turku, Finland
| |
Collapse
|
9
|
Vong CT, Tan D, Liao F, Chen Z, Chen Z, Tseng HHL, Cheang WS, Wang S, Wang Y. Ginsenoside Rk1 Ameliorates ER Stress-Induced Apoptosis through Directly Activating IGF-1R in Mouse Pancreatic [Formula: see text]-Cells and Diabetic Pancreas. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:1195-1211. [PMID: 38798150 DOI: 10.1142/s0192415x24500484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Hyperglycemia induces chronic stresses, such as oxidative stress and endoplasmic reticulum (ER) stress, which can result in [Formula: see text]-cell dysfunction and development of Type 2 Diabetes Mellitus (T2DM). Ginsenoside Rk1 is a minor ginsenoside isolated from Ginseng. It has been shown to exert anti-cancer, anti-inflammatory, anti-oxidant, and neuroprotective effects; however, its effects on pancreatic cells in T2DM have never been studied. This study aims to examine the novel effects of Ginsenoside Rk1 on ER stress-induced apoptosis in a pancreatic [Formula: see text]-cell line MIN6 and HFD-induced diabetic pancreas, and their underlying mechanisms. We demonstrated that Ginsenoside Rk1 alleviated ER stress-induced apoptosis in MIN6 cells, which was accomplished by directly targeting and activating insulin-like growth factor 1 receptor (IGF-1R), thus activating the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/Bcl-2-associated agonist of cell death (Bad)-B-cell lymphoma-2 (Bcl-2) pathway. This pathway was also confirmed in an HFD-induced diabetic pancreas. Meanwhile, the use of the IGF-1R inhibitor PQ401 abolished this anti-apoptotic effect, confirming the role of IGF-1R in mediating anti-apoptosis effects exerted by Ginsenoside Rk1. Besides, Ginsenoside Rk1 reduced pancreas weights and increased pancreatic insulin contents, suggesting that it could protect the pancreas from HFD-induced diabetes. Taken together, our study provided novel protective effects of Ginsenoside Rk1 on ER stress-induced [Formula: see text]-cell apoptosis and HFD-induced diabetic pancreases, as well as its direct target with IGF-1R, indicating that Ginsenoside Rk1 could be a potential drug for the treatment of T2DM.
Collapse
Affiliation(s)
- Chi Teng Vong
- Macau Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, P. R. China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, P. R. China
| | - Dechao Tan
- Macau Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, P. R. China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, P. R. China
| | - Fengyun Liao
- Macau Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, P. R. China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, P. R. China
| | - Zhejie Chen
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nuclei Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P. R. China
| | - Zhangmei Chen
- Macau Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, P. R. China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, P. R. China
| | - Hisa Hui Ling Tseng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, P. R. China
| | - Wai San Cheang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, P. R. China
| | - Shengpeng Wang
- Macau Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, P. R. China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, P. R. China
| | - Yitao Wang
- Macau Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, P. R. China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, P. R. China
| |
Collapse
|
10
|
Voigt JH, Lauritsen KM, Pedersen SB, Hansen TK, Møller N, Jessen N, Laurenti MC, Dalla Man C, Vella A, Gormsen LC, Søndergaard E. Four weeks SGLT2 inhibition improves beta cell function and glucose tolerance without affecting muscle free fatty acid or glucose uptake in subjects with type 2 diabetes. Basic Clin Pharmacol Toxicol 2024; 134:643-656. [PMID: 38409617 DOI: 10.1111/bcpt.13991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 01/25/2024] [Accepted: 02/06/2024] [Indexed: 02/28/2024]
Abstract
AIMS Sodium glucose co-transporter-2 (SGLT2) inhibition lowers glucose levels independently of insulin, leading to reduced insulin secretion and increased lipolysis, resulting in elevated circulating free fatty acids (FFAs). While SGLT2 inhibition improves tissue insulin sensitivity, the increase in circulating FFAs could reduce insulin sensitivity in skeletal muscle and the liver. We aimed to investigate the effects of SGLT2 inhibition on substrate utilization in skeletal muscle and the liver and to measure beta-cell function and glucose tolerance. METHODS Thirteen metformin-treated individuals with type 2 diabetes were randomized to once-daily empagliflozin 25 mg or placebo for 4 weeks in a crossover design. Skeletal muscle glucose and FFA uptake together with hepatic tissue FFA uptake were measured using [18F]FDG positron emission tomography/computed tomography (PET/CT) and [11C]palmitate PET/CT. Insulin secretion and action were estimated using the oral minimal model. RESULTS Empagliflozin did not affect glucose (0.73 ± 0.30 vs. 1.16 ± 0.64, μmol/g/min p = 0.11) or FFA (0.60 ± 0.30 vs. 0.56 ± 0.3, μmol/g/min p = 0.54) uptake in skeletal muscle. FFA uptake in the liver (21.2 ± 10.1 vs. 19 ± 8.8, μmol/100 ml/min p = 0.32) was unaffected. Empagliflozin increased total beta-cell responsivity (20 ± 8 vs. 14 ± 9, 10-9 min-1, p < 0.01) and glucose effectiveness (2.6 × 10-2 ± 0.3 × 10-2 vs. 2.4 × 10-2 ± 0.3 × 10-2, dL/kg/min, p = 0.02). CONCLUSIONS Despite improved beta-cell function and glucose tolerance, empagliflozin does not appear to affect skeletal muscle FFA or glucose uptake.
Collapse
Affiliation(s)
| | - Katrine M Lauritsen
- Steno Diabetes Center Aarhus, Aarhus, Denmark
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
- Danish Diabetes Academy, Odense University Hospital, Odense, Denmark
| | - Steen Bønløkke Pedersen
- Steno Diabetes Center Aarhus, Aarhus, Denmark
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | | | - Niels Møller
- Steno Diabetes Center Aarhus, Aarhus, Denmark
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Niels Jessen
- Steno Diabetes Center Aarhus, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Marcello C Laurenti
- Endocrine Research Unit, Department of Endocrinology, Diabetes and Nutrition, Mayo Clinic, Rochester, Minnesota, USA
| | - Chiara Dalla Man
- Department of Information Engineering, University of Padua, Padua, Italy
| | - Adrian Vella
- Endocrine Research Unit, Department of Endocrinology, Diabetes and Nutrition, Mayo Clinic, Rochester, Minnesota, USA
| | - Lars C Gormsen
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Esben Søndergaard
- Steno Diabetes Center Aarhus, Aarhus, Denmark
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
- Danish Diabetes Academy, Odense University Hospital, Odense, Denmark
- Endocrine Research Unit, Department of Endocrinology, Diabetes and Nutrition, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
11
|
Chen S, Xi M, Gao F, Li M, Dong T, Geng Z, Liu C, Huang F, Wang J, Li X, Wei P, Miao F. Evaluation of mulberry leaves’ hypoglycemic properties and hypoglycemic mechanisms. Front Pharmacol 2023; 14:1045309. [PMID: 37089923 PMCID: PMC10117911 DOI: 10.3389/fphar.2023.1045309] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 03/31/2023] [Indexed: 04/08/2023] Open
Abstract
The effectiveness of herbal medicine in treating diabetes has grown in recent years, but the precise mechanism by which it does so is still unclear to both medical professionals and diabetics. In traditional Chinese medicine, mulberry leaf is used to treat inflammation, colds, and antiviral illnesses. Mulberry leaves are one of the herbs with many medicinal applications, and as mulberry leaf study grows, there is mounting evidence that these leaves also have potent anti-diabetic properties. The direct role of mulberry leaf as a natural remedy in the treatment of diabetes has been proven in several studies and clinical trials. However, because mulberry leaf is a more potent remedy for diabetes, a deeper understanding of how it works is required. The bioactive compounds flavonoids, alkaloids, polysaccharides, polyphenols, volatile oils, sterols, amino acids, and a variety of inorganic trace elements and vitamins, among others, have been found to be abundant in mulberry leaves. Among these compounds, flavonoids, alkaloids, polysaccharides, and polyphenols have a stronger link to diabetes. Of course, trace minerals and vitamins also contribute to blood sugar regulation. Inhibiting alpha glucosidase activity in the intestine, regulating lipid metabolism in the body, protecting pancreatic -cells, lowering insulin resistance, accelerating glucose uptake by target tissues, and improving oxidative stress levels in the body are some of the main therapeutic properties mentioned above. These mechanisms can effectively regulate blood glucose levels. The therapeutic effects of the bioactive compounds found in mulberry leaves on diabetes mellitus and their associated molecular mechanisms are the main topics of this paper’s overview of the state of the art in mulberry leaf research for the treatment of diabetes mellitus.
Collapse
Affiliation(s)
- Sikai Chen
- Shaanxi University of Chinese Medicine, Xianyang, China
| | - Miaomiao Xi
- The Second Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
- Xi’an TANK Medicinal Biology Institute, Xi’an, China
| | - Feng Gao
- Shaanxi University of Chinese Medicine, Xianyang, China
| | - Min Li
- Shaanxi University of Chinese Medicine, Xianyang, China
| | - TaiWei Dong
- Shaanxi University of Chinese Medicine, Xianyang, China
| | - Zhixin Geng
- Shaanxi University of Chinese Medicine, Xianyang, China
| | - Chunyu Liu
- Shaanxi University of Chinese Medicine, Xianyang, China
| | - Fengyu Huang
- Shaanxi University of Chinese Medicine, Xianyang, China
| | - Jing Wang
- Shaanxi University of Chinese Medicine, Xianyang, China
| | - Xingyu Li
- Shaanxi University of Chinese Medicine, Xianyang, China
| | - Peifeng Wei
- Shaanxi University of Chinese Medicine, Xianyang, China
- The Second Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
- *Correspondence: Peifeng Wei, ; Feng Miao,
| | - Feng Miao
- The Second Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
- *Correspondence: Peifeng Wei, ; Feng Miao,
| |
Collapse
|
12
|
Kohara Y, Ikai S, Yoshihara A, Murao K, Sugiyama Y. Effect of chronic exposure to ketohexoses on pancreatic β-cell function in INS-1 rat insulinoma cells. Biosci Biotechnol Biochem 2023; 87:163-170. [PMID: 36413460 DOI: 10.1093/bbb/zbac190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 11/16/2022] [Indexed: 11/23/2022]
Abstract
Glucotoxicity, impaired insulin secretion, suppression of insulin gene expression, and apoptosis, in pancreatic β-cells caused by chronic hyperglycemia is a key component of the pathogenesis of type 2 diabetes. Recently, it has been reported that rare sugar d-allulose has antihyperglycemic and antihyperlipidemic effects in diabetic rats. However, the direct effects of rare sugars including d-allulose on pancreatic β-cell function are unclear. In this study, we investigated whether chronic exposure to ketohexoses causes glucotoxicity, suppression of insulin gene expression, and apoptosis, in INS-1 rat pancreatic insulinoma cells. d-Fructose, d-tagatose, l-allulose, and l-sorbose treatment for 1-week reduced insulin gene expression, whereas d-allulose, d-sorbose, l-fructose, and l-tagatose did not. All ketohexoses were transported into INS-1 cells, but were not metabolized. In addition, the ketohexoses did not induce apoptosis and did not affect glucose metabolism. These results suggest that long-term administration of d-allulose, d-sorbose, l-fructose, and l-tagatose does not affect pancreatic β-cell function.
Collapse
Affiliation(s)
- Yuri Kohara
- Department of Applied Biological Science, Faculty of Agriculture, Kagawa University, Miki, Kagawa, Japan
| | - Shuta Ikai
- Department of Applied Biological Science, Faculty of Agriculture, Kagawa University, Miki, Kagawa, Japan
| | - Akihide Yoshihara
- International Institute of Rare Sugar Research and Education, Kagawa University, Miki, Kagawa, Japan
| | - Koji Murao
- Department of Endocrinology and Metabolism, Faculty of Medicine, Kagawa University, Miki, Kagawa, Japan
| | - Yasunori Sugiyama
- Department of Applied Biological Science, Faculty of Agriculture, Kagawa University, Miki, Kagawa, Japan
| |
Collapse
|
13
|
Sajadimajd S, Deravi N, Forouhar K, Rahimi R, Kheirandish A, Bahramsoltani R. Endoplasmic reticulum as a therapeutic target in type 2 diabetes: Role of phytochemicals. Int Immunopharmacol 2023; 114:109508. [PMID: 36495694 DOI: 10.1016/j.intimp.2022.109508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 11/23/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is a metabolic disorders characterized by insulin resistance and β-cell dysfunction with an increasing worldwide incidence. Several studies have revealed that long-term glucotoxicity results in β-cell failure and death through induction of endoplasmic reticulum (ER) stress. Owing to the chronic progression of T2DM and the low effectiveness of antidiabetic drugs in long-term use, medicinal plants and their secondary metabolites seem to be the promising alternatives. Here we have provided a comprehensive review regarding the role of phytochemicals to alleviate ER stress in T2DM. Ginsenoside compound K, baicalein, quercetin, isopulegol, kaempferol, liquiritigenin, aspalathin, and tyrosol have demonstrated remarkable improvement of T2DM via modulation of ER stress. Arctigenin and total glycosides of peony have been shown to be effective in the treatment of diabetic retinopathy through modulation of ER stress. The effectiveness of grape seed proanthocyanidins and wolfberry is also shown in the relief of diabetic neuropathy and retinopathy. Resveratrol is involved in the prevention of atherosclerosis via ER stress modulation. Taken together, the data described herein revealed the capability of herbal constituents to prevent different complications of T2DM via a decrease in ER stress which open new doors to the treatment of diabetes.
Collapse
Affiliation(s)
- Soraya Sajadimajd
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| | - Niloofar Deravi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kimia Forouhar
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Roja Rahimi
- Derpartment of Traditional Pharmacy, School of Persian Medicine, Tehran University of Medical Sciences, Tehran, Iran; PhytoPharmacology Interest Group (PPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Ali Kheirandish
- Department of Pharmacology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Roodabeh Bahramsoltani
- Derpartment of Traditional Pharmacy, School of Persian Medicine, Tehran University of Medical Sciences, Tehran, Iran; PhytoPharmacology Interest Group (PPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
14
|
Study of relationship between glucagon level, glycemic status, and β-cell function in newly diagnosed T2DM patients, treated with insulin. Int J Diabetes Dev Ctries 2022. [DOI: 10.1007/s13410-022-01154-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
15
|
ZnT8 loss-of-function accelerates functional maturation of hESC-derived β cells and resists metabolic stress in diabetes. Nat Commun 2022; 13:4142. [PMID: 35842441 PMCID: PMC9288460 DOI: 10.1038/s41467-022-31829-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 07/04/2022] [Indexed: 12/21/2022] Open
Abstract
Human embryonic stem cell-derived β cells (SC-β cells) hold great promise for treatment of diabetes, yet how to achieve functional maturation and protect them against metabolic stresses such as glucotoxicity and lipotoxicity remains elusive. Our single-cell RNA-seq analysis reveals that ZnT8 loss of function (LOF) accelerates the functional maturation of SC-β cells. As a result, ZnT8 LOF improves glucose-stimulated insulin secretion (GSIS) by releasing the negative feedback of zinc inhibition on insulin secretion. Furthermore, we demonstrate that ZnT8 LOF mutations endow SC-β cells with resistance to lipotoxicity/glucotoxicity-triggered cell death by alleviating endoplasmic reticulum (ER) stress through modulation of zinc levels. Importantly, transplantation of SC-β cells with ZnT8 LOF into mice with preexisting diabetes significantly improves glycemia restoration and glucose tolerance. These findings highlight the beneficial effect of ZnT8 LOF on the functional maturation and survival of SC-β cells that are useful as a potential source for cell replacement therapies. Immature function and fragility hinder application of hESC-derived β cells (SC-β cell) for diabetes cell therapy. Here, the authors identify ZnT8 as a gene editing target to enhance the insulin secretion and cell survival under metabolic stress by abolishing zinc transport in SC-β cells.
Collapse
|
16
|
Uchihara M, Bouchi R, Kodani N, Saito S, Miyazato Y, Umamoto K, Sugimoto H, Kobayashi M, Hikida S, Akiyama Y, Ihana‐Sugiyama N, Ohsugi M, Tanabe A, Ueki K, Takasaki J, Hojo M, Kajio H. Impact of newly diagnosed diabetes on coronavirus disease 2019 severity and hyperglycemia. J Diabetes Investig 2022; 13:1086-1093. [PMID: 35075818 PMCID: PMC9153833 DOI: 10.1111/jdi.13754] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/11/2022] [Accepted: 01/19/2022] [Indexed: 01/08/2023] Open
Abstract
AIMS/INTRODUCTION Diabetes is associated with poor clinical outcomes of coronavirus disease 2019 (COVID-19). However, the impact of newly diagnosed diabetes on prognosis has not been clarified. The objective of this study was to show the features and outcome of COVID-19 patients with newly diagnosed diabetes in Japan. MATERIALS AND METHODS We retrospectively analyzed 62 patients with diabetes hospitalized for COVID-19 between 1 April and 18 August 2021 at the National Center for Global Health and Medicine in Tokyo, Japan. We evaluated the worst severity of COVID-19 and plasma blood glucose levels in patients with newly diagnosed diabetes or pre-existing diabetes. RESULTS This study included 62 confirmed COVID-19 patients with diabetes, including 19 (30.6%) patients with newly diagnosed diabetes and 43 (69.4%) patients with pre-existing diabetes. Patients with newly diagnosed diabetes significantly progressed to a critical condition more frequently during hospitalization than patients with pre-existing diabetes (52.6% vs 20.9%, P = 0.018). In addition, patients with newly diagnosed diabetes had significantly higher average plasma blood glucose levels for the first 3 days after admission than those with pre-existing diabetes. CONCLUSIONS Our study suggests that the proportion of COVID-19 patients who are newly diagnosed with diabetes is high, and they have an increased risk of developing severe disease than those with pre-existing diabetes. It might be advisable that at the point of COVID-19 diagnosis, blood glucose and glycated hemoglobin levels be assessed in all patients.
Collapse
Affiliation(s)
- Masaki Uchihara
- Department of Diabetes, Endocrinology and MetabolismNational Center for Global Health and MedicineTokyoJapan
| | - Ryotaro Bouchi
- Department of Diabetes, Endocrinology and MetabolismNational Center for Global Health and MedicineTokyoJapan
- Diabetes and Metabolism Information CenterResearch InstituteNational Center for Global Health and MedicineTokyoJapan
| | - Noriko Kodani
- Department of Diabetes, Endocrinology and MetabolismNational Center for Global Health and MedicineTokyoJapan
| | - Sho Saito
- Disease Control and Prevention CenterNational Center for Global Health and MedicineTokyoJapan
| | - Yusuke Miyazato
- Disease Control and Prevention CenterNational Center for Global Health and MedicineTokyoJapan
| | - Kotaro Umamoto
- Department of Diabetes, Endocrinology and MetabolismNational Center for Global Health and MedicineTokyoJapan
| | - Hirofumi Sugimoto
- Department of Diabetes, Endocrinology and MetabolismNational Center for Global Health and MedicineTokyoJapan
| | - Michi Kobayashi
- Department of Diabetes, Endocrinology and MetabolismNational Center for Global Health and MedicineTokyoJapan
| | - Sayaka Hikida
- Disease Control and Prevention CenterNational Center for Global Health and MedicineTokyoJapan
| | - Yutaro Akiyama
- Disease Control and Prevention CenterNational Center for Global Health and MedicineTokyoJapan
| | - Noriko Ihana‐Sugiyama
- Department of Diabetes, Endocrinology and MetabolismNational Center for Global Health and MedicineTokyoJapan
- Diabetes and Metabolism Information CenterResearch InstituteNational Center for Global Health and MedicineTokyoJapan
| | - Mitsuru Ohsugi
- Department of Diabetes, Endocrinology and MetabolismNational Center for Global Health and MedicineTokyoJapan
- Diabetes and Metabolism Information CenterResearch InstituteNational Center for Global Health and MedicineTokyoJapan
| | - Akiyo Tanabe
- Department of Diabetes, Endocrinology and MetabolismNational Center for Global Health and MedicineTokyoJapan
| | - Kohjiro Ueki
- Department of Diabetes, Endocrinology and MetabolismNational Center for Global Health and MedicineTokyoJapan
- Department of Molecular Diabetic MedicineDiabetes Research CenterResearch InstituteNational Center for Global Health and MedicineTokyoJapan
| | - Jin Takasaki
- Disease Control and Prevention CenterNational Center for Global Health and MedicineTokyoJapan
- Department of Respiratory MedicineNational Center for Global Health and MedicineTokyoJapan
| | - Masayuki Hojo
- Department of Respiratory MedicineNational Center for Global Health and MedicineTokyoJapan
| | - Hiroshi Kajio
- Department of Diabetes, Endocrinology and MetabolismNational Center for Global Health and MedicineTokyoJapan
| |
Collapse
|
17
|
Shanak S, Bassalat N, Barghash A, Kadan S, Ardah M, Zaid H. Drug Discovery of Plausible Lead Natural Compounds That Target the Insulin Signaling Pathway: Bioinformatics Approaches. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:2832889. [PMID: 35356248 PMCID: PMC8958086 DOI: 10.1155/2022/2832889] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/16/2022] [Accepted: 02/09/2022] [Indexed: 12/11/2022]
Abstract
The growing smooth talk in the field of natural compounds is due to the ancient and current interest in herbal medicine and their potentially positive effects on health. Dozens of antidiabetic natural compounds were reported and tested in vivo, in silico, and in vitro. The role of these natural compounds, their actions on the insulin signaling pathway, and the stimulation of the glucose transporter-4 (GLUT4) insulin-responsive translocation to the plasma membrane (PM) are all crucial in the treatment of diabetes and insulin resistance. In this review, we collected and summarized a group of available in vivo and in vitro studies which targeted isolated phytochemicals with possible antidiabetic activity. Moreover, the in silico docking of natural compounds with some of the insulin signaling cascade key proteins is also summarized based on the current literature. In this review, hundreds of recent studies on pure natural compounds that alleviate type II diabetes mellitus (type II DM) were revised. We focused on natural compounds that could potentially regulate blood glucose and stimulate GLUT4 translocation through the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway. On attempt to point out potential new natural antidiabetic compounds, this review also focuses on natural ingredients that were shown to interact with proteins in the insulin signaling pathway in silico, regardless of their in vitro/in vivo antidiabetic activity. We invite interested researchers to test these compounds as potential novel type II DM drugs and explore their therapeutic mechanisms.
Collapse
Affiliation(s)
- Siba Shanak
- Faculty of Sciences, Arab American University, P.O Box 240, Jenin, State of Palestine
| | - Najlaa Bassalat
- Faculty of Sciences, Arab American University, P.O Box 240, Jenin, State of Palestine
- Faculty of Medicine, Arab American University, P.O Box 240, Jenin, State of Palestine
| | - Ahmad Barghash
- Computer Science Department, German Jordanian University, Madaba Street. P.O. Box 35247, Amman 11180, Jordan
| | - Sleman Kadan
- Qasemi Research Center, Al-Qasemi Academic College, P.O Box 124, Baqa El-Gharbia 30100, Israel
| | - Mahmoud Ardah
- Faculty of Sciences, Arab American University, P.O Box 240, Jenin, State of Palestine
| | - Hilal Zaid
- Faculty of Medicine, Arab American University, P.O Box 240, Jenin, State of Palestine
- Qasemi Research Center, Al-Qasemi Academic College, P.O Box 124, Baqa El-Gharbia 30100, Israel
| |
Collapse
|
18
|
Longo M, Scappaticcio L, Cirillo P, Maio A, Carotenuto R, Maiorino MI, Bellastella G, Esposito K. Glycemic Control and the Heart: The Tale of Diabetic Cardiomyopathy Continues. Biomolecules 2022; 12:biom12020272. [PMID: 35204778 PMCID: PMC8961546 DOI: 10.3390/biom12020272] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 01/25/2022] [Accepted: 02/01/2022] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases are the leading cause of death in people with diabetes. Diabetic cardiomyopathy (DC) is an important complication of diabetes and represents a distinct subtype of heart failure that occurs in absence of cardiovascular diseases. Chronic hyperglycemia and hyperinsulinemia along with insulin resistance and inflammatory milieu are the main mechanisms involved in the pathophysiology of DC. Changes in lifestyle favoring healthy dietary patterns and physical activity, combined with more innovative anti-diabetes therapies, are the current treatment strategies to safeguard the cardiovascular system. This review aims at providing an updated comprehensive overview of clinical, pathogenetic, and molecular aspects of DC, with a focus on the effects of anti-hyperglycemic drugs on the prevention of pump dysfunction and consequently on cardiovascular health in type 2 diabetes.
Collapse
Affiliation(s)
- Miriam Longo
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.L.); (L.S.); (P.C.); (A.M.); (R.C.); (M.I.M.); (G.B.)
- Division of Endocrinology and Metabolic Diseases, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Lorenzo Scappaticcio
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.L.); (L.S.); (P.C.); (A.M.); (R.C.); (M.I.M.); (G.B.)
| | - Paolo Cirillo
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.L.); (L.S.); (P.C.); (A.M.); (R.C.); (M.I.M.); (G.B.)
| | - Antonietta Maio
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.L.); (L.S.); (P.C.); (A.M.); (R.C.); (M.I.M.); (G.B.)
| | - Raffaela Carotenuto
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.L.); (L.S.); (P.C.); (A.M.); (R.C.); (M.I.M.); (G.B.)
- Division of Endocrinology and Metabolic Diseases, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Maria Ida Maiorino
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.L.); (L.S.); (P.C.); (A.M.); (R.C.); (M.I.M.); (G.B.)
- Division of Endocrinology and Metabolic Diseases, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Giuseppe Bellastella
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.L.); (L.S.); (P.C.); (A.M.); (R.C.); (M.I.M.); (G.B.)
- Division of Endocrinology and Metabolic Diseases, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Katherine Esposito
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.L.); (L.S.); (P.C.); (A.M.); (R.C.); (M.I.M.); (G.B.)
- Division of Endocrinology and Metabolic Diseases, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
- Correspondence: ; Tel.: +39-08-156-65031
| |
Collapse
|
19
|
Liu ZH, Li B. Chlorogenic acid and β-glucan from highland barley grain ameliorate β-cell dysfunction via inhibiting apoptosis and improving cell proliferation. Food Funct 2021; 12:10040-10052. [PMID: 34515712 DOI: 10.1039/d1fo01532j] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Recent studies have reported that highland barley as a whole grain diet has anti-hyperglycemic effects, however little information is available about the active compounds that ameliorate pancreatic β-cell dysfunction and the related mechanisms. In this study, chlorogenic acid (CA) and β-glucan (BG) were identified as the active compounds that ameliorated β-cell dysfunction. CA ameliorated β-cell dysfunction by inhibiting cell apoptosis and improving glucose-stimulated insulin secretion via targeting G protein-coupled receptor 40 (GPR40) and regulating the phospholipase C β (PLCβ) pathway. BG ameliorated β-cell dysfunction by improving cell proliferation via targeting mammalian target of rapamycin (mTOR) and regulating the protein kinase B (Akt)/glycogen synthase kinase-3β (GSK-3β) pathway. Furthermore, CA and BG improved β-cell sensitivity and pancreatic insulin secretion, and inhibited β-cell apoptosis in impaired glucose tolerance (IGT) mice. Notably, CA restored homeostasis model assessment (HOMA)-β values and Ca2+-ATP and K+-ATP levels back to normal levels, and BG at 300 mg per kg BW restored β-cell insulin contents back to normal levels in IGT mice. Additionally, the combination of CA and BG had an additive effect on ameliorating β-cell dysfunction. These results help develop whole highland barley grain as a functional food for preventing type 2 diabetes by ameliorating pancreatic β-cell damage.
Collapse
Affiliation(s)
- Ze-Hua Liu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| | - Bo Li
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China. .,Key Laboratory of Functional Dairy, Ministry of Education, Beijing 100083, China
| |
Collapse
|
20
|
Bonnet F, Chen H, Cooper A, Gomes MB, Ji L, Leigh P, Ramirez L, Shestakova MV, Shimomura I, Siddiqui A, Tang F, Vora J, Watada H, Khunti K. What are the factors associated with long-term glycaemic control in patients with type 2 diabetes and elevated glycated haemoglobin (≥7.0%) at initiation of second-line therapy? Results from the DISCOVER study. Diabetes Obes Metab 2021; 23:2336-2343. [PMID: 34212481 DOI: 10.1111/dom.14476] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/11/2021] [Accepted: 06/27/2021] [Indexed: 11/26/2022]
Abstract
AIMS Glycaemic control is a cornerstone of type 2 diabetes (T2D) management. We assessed factors associated with good long-term glycaemic control in patients with glycated haemoglobin (HbA1c) ≥7.0% at initiation of second-line glucose-lowering therapy, using data from DISCOVER, a global, prospective, 3-year observational study of patients with T2D. MATERIALS AND METHODS This analysis included patients with HbA1c ≥7.0% at baseline (initiation of second-line therapy). Multivariable regression models assessed factors associated with having HbA1c <7.0% at 3 years in two distinct groups: patients with (a) HbA1c ≥7.0% and <9.0%, and (b) HbA1c ≥9.0% at baseline. RESULTS In total, 7575 patients with baseline HbA1c ≥7.0% were included (2233 with baseline HbA1c ≥9.0%). At 6 months, 43.7% and 24.2% of patients had an HbA1c level <7.0% in groups a and b, respectively; the corresponding proportions at 3 years were 45.8% and 29.3%. Having HbA1c <7.0% at 6 months (vs. ≥7.0%) was the strongest predictor of having HbA1c <7.0% at 3 years in both group a and group b [odds ratio (95% confidence interval): 2.01 (1.77-2.27) and 2.68 (2.10-3.41), respectively]. Longer T2D duration was associated with a decreased likelihood of having HbA1c <7.0% at 3 years. CONCLUSIONS In patients with poor glycaemic control at initiation of second-line therapy, early attainment of HbA1c <7.0% appears predictive of long-term glycaemic control, suggesting that timely modification of treatment strategies in patients with elevated HbA1c after 6 months is important to minimize therapeutic inertia.
Collapse
Affiliation(s)
- Fabrice Bonnet
- Department of Endocrinology, Rennes University Hospital, Rennes 1 University, Rennes, France
| | | | | | | | - Linong Ji
- Peking University People's Hospital, Beijing, China
| | | | | | - Marina V Shestakova
- Endocrinology Research Center, Diabetes Institute, Moscow, Russian Federation
| | | | | | - Fengming Tang
- Saint Luke's Mid America Heart Institute, Kansas City, Missouri, USA
| | | | - Hirotaka Watada
- Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | | |
Collapse
|
21
|
Pereira APA, Fernando Figueiredo Angolini C, de Souza-Sporkens JC, da Silva TA, Coutinho Franco de Oliveira H, Pastore GM. Brazilian sunberry (Solanum oocarpum Sendtn): Alkaloid composition and improvement of mitochondrial functionality and insulin secretion of INS-1E cells. Food Res Int 2021; 148:110589. [PMID: 34507734 DOI: 10.1016/j.foodres.2021.110589] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 05/24/2021] [Accepted: 07/02/2021] [Indexed: 11/30/2022]
Abstract
Chronic high-glucose levels induce the generation of reactive oxygen species leading to mitochondrial dysfunction, which is one of the pathological triggers in the development of diabetes. This study investigated the alkaloid composition of two fruits of the genus Solanum, fruta-do-lobo (Solanum lycocarpum) and juá-açu (Solanum oocarpum), and their capacity to protect against oxidative damage and defective insulin secretion induced by chronic high-glucose levels. LC-MS and molecular network of fruit crude extracts reveals that juá-açu and fruta-do-lobo contain kukoamines and glycoalkaloids, respectively. Two purification processes were used to enrich those alkaloids. Fruta-do-lobo extract rich in glycoalkaloids showed a strong cytotoxicity effect, however the juá-açu enriched extract was able to protect mitochondrial functionality against glucotoxicity and stimulate insulin secretion even under conditions of hyperglycemia. These results are promising and suggest that juá-açu is a potential source of bioactive compounds for adjuvant/co-adjuvant therapy for diabetes.
Collapse
Affiliation(s)
- Ana Paula Aparecida Pereira
- Department of Food Science, Faculty of Food Engineering, University of Campinas (UNICAMP), Rua Monteiro Lobato, 80, Campinas, São Paulo 13083-862, Brazil; Department of Food and Nutrition, Faculty of Nutrition, Federal University of Mato Grosso, Cuiabá, Mato Grosso, Brazil.
| | | | - Jane Cristina de Souza-Sporkens
- Department of Structural and Functional Biology, Biology Institute, University of Campinas, Rua Monteiro Lobato, 255, Campinas, São Paulo 13083-862, Brazil
| | - Tomaz Antonio da Silva
- Center for Natural and Human Sciences, University of ABC (UFABC), Santo André, São Paulo, Brazil
| | - Helena Coutinho Franco de Oliveira
- Department of Structural and Functional Biology, Biology Institute, University of Campinas, Rua Monteiro Lobato, 255, Campinas, São Paulo 13083-862, Brazil
| | - Glaucia Maria Pastore
- Department of Food Science, Faculty of Food Engineering, University of Campinas (UNICAMP), Rua Monteiro Lobato, 80, Campinas, São Paulo 13083-862, Brazil
| |
Collapse
|
22
|
Nakane T, Matsumoto S, Iida S, Ido A, Fukunaga K, Murao K, Sugiyama Y. Candidate plasticity gene 16 and jun dimerization protein 2 are involved in the suppression of insulin gene expression in rat pancreatic INS-1 β-cells. Mol Cell Endocrinol 2021; 527:111240. [PMID: 33676985 DOI: 10.1016/j.mce.2021.111240] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/24/2021] [Accepted: 03/01/2021] [Indexed: 11/22/2022]
Abstract
Chronic hyperglycemia causes pancreatic β-cell dysfunction, impaired insulin secretion and the suppression of insulin gene expression. This phenomenon is referred to as glucotoxicity, and is a critical component of the pathogenesis of type 2 diabetes. We previously reported that the expression of candidate plasticity gene 16 (CPG16) was higher in rat pancreatic INS-1 β-cells under glucotoxic conditions and CPG16 suppressed insulin promoter activity. However, the molecular mechanisms of the CPG16-mediated suppression of insulin gene expression are unclear. In this study, we found that CPG16 directly bound and phosphorylated jun dimerization protein 2 (JDP2), an AP-1 family transcription factor. CPG16 co-localized with JDP2 in the nucleus of INS-1 cells. JDP2 bound to the G1 element of the insulin promoter and up-regulated promoter activity. Finally, CPG16 suppressed the up-regulation of insulin promoter activity by JDP2 in a kinase activity-dependent manner. These results suggest that CPG16 suppresses insulin promoter activity by phosphorylating JDP2.
Collapse
Affiliation(s)
- Tatsuto Nakane
- Department of Life Sciences, Faculty of Agriculture, Kagawa University, Kagawa, Japan
| | - Suzuka Matsumoto
- Department of Life Sciences, Faculty of Agriculture, Kagawa University, Kagawa, Japan
| | - Satoshi Iida
- Department of Life Sciences, Faculty of Agriculture, Kagawa University, Kagawa, Japan
| | - Ayae Ido
- Department of Life Sciences, Faculty of Agriculture, Kagawa University, Kagawa, Japan
| | - Kensaku Fukunaga
- Department of Endocrinology and Metabolism, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Koji Murao
- Department of Endocrinology and Metabolism, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Yasunori Sugiyama
- Department of Life Sciences, Faculty of Agriculture, Kagawa University, Kagawa, Japan.
| |
Collapse
|
23
|
Wang Z, Zhao L, He S. Triglyceride-glucose index as predictor for future type 2 diabetes mellitus in a Chinese population in southwest China: a 15-year prospective study. Endocrine 2021; 72:124-131. [PMID: 33433893 DOI: 10.1007/s12020-020-02589-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 12/09/2020] [Indexed: 02/05/2023]
Abstract
PURPOSE Triglyceride-glucose (TyG) index is an emerging surrogate predictor of incident type 2 diabetes mellitus (T2DM). The study aimed to examine the association between TyG index and incident T2DM in a prospective Chinese cohort. METHODS The data were collected in 1992 and recollected in 2007 from the same group of 687 participants. The association between TyG index and T2DM was analysed. RESULTS During follow-up, 74 participants developed T2DM and the risk of T2DM increased with TyG index. The adjusted hazard ratio (HR) was 3.36 (95% CI: 1.52-7.39, P < 0.001) comparing the top TyG quartile to the bottom quartile. Smooth curve fitting revealed a nonlinear association and threshold effect between TyG index and incident T2DM with a nadir of risk when TyG index was around 8.51. For TyG ≤ 8.51, the risk of incident T2DM tended to decrease with per SD increase in TyG but no statistical significance was achieved (adjusted HR: 0.69, 95% CI: 0.43-1.12, P = 0.133). For TyG > 8.51, the risk of incident T2DM significantly increased by 38% with per SD increase in TyG (adjusted HR: 1.38, 95% CI: 1.14-1.67, P = 0.001). Time-dependent receiver operating curve suggested helpful discriminative power of TyG index for T2DM. It also significantly promoted the reclassification ability beyond the baseline risk model with net reclassification index of 0.159 (P = 0.020). Sensitivity analysis excluding participants with prediabetes demonstrated similar results. CONCLUSIONS The TyG index was a significant and independent predictor for future T2DM development. The shape of relationship will require further studies.
Collapse
Affiliation(s)
- Ziqiong Wang
- Department of Cardiology, West China Hospital of Sichuan University, Chengdu, China
| | - Liming Zhao
- Department of Cardiovascular Medicine, Hospital of Chengdu Office of People's Government of Tibet Autonomous Region, Chengdu, China
| | - Sen He
- Department of Cardiology, West China Hospital of Sichuan University, Chengdu, China.
| |
Collapse
|
24
|
Koyama R, Ookawara M, Watanabe M, Moritoh Y. Chronic Exposure to SCO-267, an Allosteric GPR40 Full Agonist, Is Effective in Improving Glycemic Control in Rats. Mol Pharmacol 2021; 99:286-293. [PMID: 33547250 DOI: 10.1124/molpharm.120.000168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 01/15/2021] [Indexed: 11/22/2022] Open
Abstract
Full agonist-mediated activation of free fatty acid receptor 1 (FFAR1/GPR40) alleviates diabetes in rodents. Considering that diabetes is a chronic disease, assessment of treatment durability of chronic exposure to a GPR40 full agonist is pivotal for treating patients with diabetes. However, the physiologic significance of chronic in vitro and in vivo exposure to GPR40 full agonists is largely unclear. Here, we evaluated the in vitro and in vivo effects of chronic treatment with SCO-267, a GPR40 full agonist, on signal transduction and glucose control. In vitro experiments showed that SCO-267 is an allosteric full agonist for GPR40, which activates the Gα q, Gα s, and Gα 12/13 pathways and β-arrestin recruitment. The calcium signal response was largely sustained in GPR40-overexpressing CHO cells even after prolonged incubation with SCO-267. To evaluate the in vivo relevance of chronic exposure to GPR40 full agonists, SCO-267 (1 and 10 mg/kg) was administered once daily to neonatally streptozotocin-induced diabetic rats for 15-33 days, and glucose control was evaluated. After 15 days of dosing followed by the drug washout period, SCO-267 improved glucose tolerance, most likely by increasing insulin sensitivity in rats. After 33 days, repeated exposure to SCO-267 was highly effective in improving glucose tolerance in rats. Furthermore, chronic exposure to SCO-267 increased pancreatic insulin content. These results demonstrated that even after chronic exposure, SCO-267 effectively activates GPR40 in cells and rats, suggesting the clinical application of SCO-267 in treating chronic diseases including diabetes. SIGNIFICANCE STATEMENT: GPR40 is a validated therapeutic target for diabetes. This study showed that even after chronic exposure, SCO-267, an allosteric GPR40 full agonist, effectively activates GPR40 in cells and rats; these results suggest a durable efficacy of SCO-267 in patients.
Collapse
Affiliation(s)
- Ryokichi Koyama
- Research Division, SCOHIA PHARMA, Inc., Fujisawa, Kanagawa, Japan
| | - Mitsugi Ookawara
- Research Division, SCOHIA PHARMA, Inc., Fujisawa, Kanagawa, Japan
| | | | - Yusuke Moritoh
- Research Division, SCOHIA PHARMA, Inc., Fujisawa, Kanagawa, Japan
| |
Collapse
|
25
|
Hayden MR. An Immediate and Long-Term Complication of COVID-19 May Be Type 2 Diabetes Mellitus: The Central Role of β-Cell Dysfunction, Apoptosis and Exploration of Possible Mechanisms. Cells 2020; 9:E2475. [PMID: 33202960 PMCID: PMC7697826 DOI: 10.3390/cells9112475] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/03/2020] [Accepted: 11/10/2020] [Indexed: 12/15/2022] Open
Abstract
The novel coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) was declared a pandemic by the WHO on 19 March 2020. This pandemic is associated with markedly elevated blood glucose levels and a remarkable degree of insulin resistance, which suggests pancreatic islet β-cell dysfunction or apoptosis and insulin's inability to dispose of glucose into cellular tissues. Diabetes is known to be one of the top pre-existing co-morbidities associated with the severity of COVID-19 along with hypertension, cardiocerebrovascular disease, advanced age, male gender, and recently obesity. This review focuses on how COVID-19 may be responsible for the accelerated development of type 2 diabetes mellitus (T2DM) as one of its acute and suspected long-term complications. These observations implicate an active role of metabolic syndrome, systemic and tissue islet renin-angiotensin-aldosterone system, redox stress, inflammation, islet fibrosis, amyloid deposition along with β-cell dysfunction and apoptosis in those who develop T2DM. Utilizing light and electron microscopy in preclinical rodent models and human islets may help to better understand how COVID-19 accelerates islet and β-cell injury and remodeling to result in the long-term complications of T2DM.
Collapse
Affiliation(s)
- Melvin R Hayden
- Departments of Internal Medicine, Endocrinology Diabetes and Metabolism, Diabetes and Cardiovascular Disease Center, University of Missouri-Columbia School of Medicine, Columbia, MO 65212, USA
| |
Collapse
|
26
|
Kehm R, Jähnert M, Deubel S, Flore T, König J, Jung T, Stadion M, Jonas W, Schürmann A, Grune T, Höhn A. Redox homeostasis and cell cycle activation mediate beta-cell mass expansion in aged, diabetes-prone mice under metabolic stress conditions: Role of thioredoxin-interacting protein (TXNIP). Redox Biol 2020; 37:101748. [PMID: 33128997 PMCID: PMC7589534 DOI: 10.1016/j.redox.2020.101748] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 10/02/2020] [Indexed: 12/12/2022] Open
Abstract
Overnutrition contributes to insulin resistance, obesity and metabolic stress, initiating a loss of functional beta-cells and diabetes development. Whether these damaging effects are amplified in advanced age is barely investigated. Therefore, New Zealand Obese (NZO) mice, a well-established model for the investigation of human obesity-associated type 2 diabetes, were fed a metabolically challenging diet with a high-fat, carbohydrate restricted period followed by a carbohydrate intervention in young as well as advanced age. Interestingly, while young NZO mice developed massive hyperglycemia in response to carbohydrate feeding, leading to beta-cell dysfunction and cell death, aged counterparts compensated the increased insulin demand by persistent beta-cell function and beta-cell mass expansion. Beta-cell loss in young NZO islets was linked to increased expression of thioredoxin-interacting protein (TXNIP), presumably initiating an apoptosis-signaling cascade via caspase-3 activation. In contrast, islets of aged NZOs exhibited a sustained redox balance without changes in TXNIP expression, associated with higher proliferative potential by cell cycle activation. These findings support the relevance of a maintained proliferative potential and redox homeostasis for preserving islet functionality under metabolic stress, with the peculiarity that this adaptive response emerged with advanced age in diabetes-prone NZO mice. Differential expression of redox and cell cycle genes in young and aged islets. Increased TXNIP expression is associated with the induction of beta-cell apoptosis. Islets of aged mice maintained redox homeostasis and proliferative potential. Aging under diet-induced metabolic stress does not amplify beta-cell failure.
Collapse
Affiliation(s)
- Richard Kehm
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558, Nuthetal, Germany; German Center for Diabetes Research (DZD), 85764, Muenchen-Neuherberg, Germany.
| | - Markus Jähnert
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558, Nuthetal, Germany; German Center for Diabetes Research (DZD), 85764, Muenchen-Neuherberg, Germany.
| | - Stefanie Deubel
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558, Nuthetal, Germany.
| | - Tanina Flore
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558, Nuthetal, Germany.
| | - Jeannette König
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558, Nuthetal, Germany.
| | - Tobias Jung
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558, Nuthetal, Germany; University of Potsdam, Institute of Nutritional Science, 14558, Nuthetal, Germany.
| | - Mandy Stadion
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558, Nuthetal, Germany; German Center for Diabetes Research (DZD), 85764, Muenchen-Neuherberg, Germany.
| | - Wenke Jonas
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558, Nuthetal, Germany; German Center for Diabetes Research (DZD), 85764, Muenchen-Neuherberg, Germany.
| | - Annette Schürmann
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558, Nuthetal, Germany; German Center for Diabetes Research (DZD), 85764, Muenchen-Neuherberg, Germany; University of Potsdam, Institute of Nutritional Science, 14558, Nuthetal, Germany.
| | - Tilman Grune
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558, Nuthetal, Germany; German Center for Diabetes Research (DZD), 85764, Muenchen-Neuherberg, Germany; NutriAct-Competence Cluster Nutrition Research Berlin-Potsdam, 14458, Nuthetal, Germany; German Center for Cardiovascular Research (DZHK), 10117, Berlin, Germany; University of Potsdam, Institute of Nutritional Science, 14558, Nuthetal, Germany.
| | - Annika Höhn
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558, Nuthetal, Germany; German Center for Diabetes Research (DZD), 85764, Muenchen-Neuherberg, Germany.
| |
Collapse
|
27
|
Courtney CM, Shyr ZA, Yan Z, Onufer EJ, Steinberger AE, Tecos ME, Barron LK, Guo J, Remedi MS, Warner BW. Alterations in pancreatic islet cell function in response to small bowel resection. Am J Physiol Gastrointest Liver Physiol 2020; 319:G36-G42. [PMID: 32463335 PMCID: PMC7468758 DOI: 10.1152/ajpgi.00282.2019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
After 50% proximal small bowel resection (SBR) in mice, we have demonstrated hepatic steatosis, impaired glucose metabolism without insulin resistance, and increased pancreatic islet area. We sought to determine the consequences of SBR on pancreatic β-cell morphology, proliferation, and expression of a key regulatory hormone, glucagon-like peptide-1 (GLP-1). C57BL/6 mice underwent 50% SBR or sham operation. At 10 wk, pancreatic insulin content and secretion was measured by ELISA. Immunohistochemistry was performed to determine structural alterations in pancreatic α-and β-cells. Western blot analysis was used to measure GLP-1R expression, and immunoassay was used to measure plasma insulin and GLP-1. Experiments were repeated by administering a GLP-1 agonist (exendin-4) to a cohort of mice following SBR. After SBR, there was pancreatic islet hypertrophy and impaired glucose tolerance. The proportion of α and β cells was not grossly altered. Whole pancreas and pancreatic islet insulin content was not significantly different; however, SBR mice demonstrated decreased insulin secretion in both static incubation and islet perfusion experiments. The expression of pancreatic GLP-1R was decreased approximately twofold after SBR, compared with sham and serum GLP-1, was decreased. These metabolic derangements were mitigated after administration of the GLP-1 agonist. Following massive SBR, there is significant hypertrophy of pancreatic islet cells with morphologically intact α- and β-cells. Significantly reduced pancreatic insulin release in both static and dynamic conditions demonstrate a perturbed second phase of insulin secretion. GLP-1 is a key mediator of this amplification pathway. Decreased expression of serum GLP-1 and pancreatic GLP-1R in face of no change in insulin content presents a novel pathway for enteropancreatic glucose regulation following SBR.NEW & NOTEWORTHY Metabolic changes occur following intestinal resection; however, the effects on pancreatic function are unknown. Prior studies have demonstrated that glucagon-like protein-1 (GLP-1) signaling is a crucial player in the improved insulin sensitivity after bariatric surgery. In this study, we explore the effect of massive small bowel resection on gut hormone physiology and provide novel insights into the enteropancreatic axis.
Collapse
Affiliation(s)
- Cathleen M. Courtney
- 1Division of Pediatric Surgery, Department of Surgery, St. Louis Children’s Hospital, Washington University School of Medicine, St. Louis, Missouri
| | - Zeenat A. Shyr
- 2Division of Endocrinology, Metabolism, and Lipid Research, Washington University in St. Louis, Missouri
| | - Zihan Yan
- 2Division of Endocrinology, Metabolism, and Lipid Research, Washington University in St. Louis, Missouri
| | - Emily Jean Onufer
- 1Division of Pediatric Surgery, Department of Surgery, St. Louis Children’s Hospital, Washington University School of Medicine, St. Louis, Missouri
| | - Allie E. Steinberger
- 1Division of Pediatric Surgery, Department of Surgery, St. Louis Children’s Hospital, Washington University School of Medicine, St. Louis, Missouri
| | - Maria E. Tecos
- 1Division of Pediatric Surgery, Department of Surgery, St. Louis Children’s Hospital, Washington University School of Medicine, St. Louis, Missouri
| | - Lauren K. Barron
- 1Division of Pediatric Surgery, Department of Surgery, St. Louis Children’s Hospital, Washington University School of Medicine, St. Louis, Missouri
| | - Jun Guo
- 1Division of Pediatric Surgery, Department of Surgery, St. Louis Children’s Hospital, Washington University School of Medicine, St. Louis, Missouri
| | - Maria S. Remedi
- 2Division of Endocrinology, Metabolism, and Lipid Research, Washington University in St. Louis, Missouri
| | - Brad W. Warner
- 1Division of Pediatric Surgery, Department of Surgery, St. Louis Children’s Hospital, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
28
|
Liu B, Wang Y, Zhang Y, Yan B. Mechanisms of Protective Effects of SGLT2 Inhibitors in Cardiovascular Disease and Renal Dysfunction. Curr Top Med Chem 2019; 19:1818-1849. [PMID: 31456521 DOI: 10.2174/1568026619666190828161409] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 07/09/2019] [Accepted: 07/25/2019] [Indexed: 02/07/2023]
Abstract
Type 2 diabetes mellitus is one of the most common forms of the disease worldwide. Hyperglycemia and insulin resistance play key roles in type 2 diabetes mellitus. Renal glucose reabsorption is an essential feature in glycaemic control. Kidneys filter 160 g of glucose daily in healthy subjects under euglycaemic conditions. The expanding epidemic of diabetes leads to a prevalence of diabetes-related cardiovascular disorders, in particular, heart failure and renal dysfunction. Cellular glucose uptake is a fundamental process for homeostasis, growth, and metabolism. In humans, three families of glucose transporters have been identified, including the glucose facilitators GLUTs, the sodium-glucose cotransporter SGLTs, and the recently identified SWEETs. Structures of the major isoforms of all three families were studied. Sodium-glucose cotransporter (SGLT2) provides most of the capacity for renal glucose reabsorption in the early proximal tubule. A number of cardiovascular outcome trials in patients with type 2 diabetes have been studied with SGLT2 inhibitors reducing cardiovascular morbidity and mortality. The current review article summarises these aspects and discusses possible mechanisms with SGLT2 inhibitors in protecting heart failure and renal dysfunction in diabetic patients. Through glucosuria, SGLT2 inhibitors reduce body weight and body fat, and shift substrate utilisation from carbohydrates to lipids and, possibly, ketone bodies. These pleiotropic effects of SGLT2 inhibitors are likely to have contributed to the results of the EMPA-REG OUTCOME trial in which the SGLT2 inhibitor, empagliflozin, slowed down the progression of chronic kidney disease and reduced major adverse cardiovascular events in high-risk individuals with type 2 diabetes. This review discusses the role of SGLT2 in the physiology and pathophysiology of renal glucose reabsorption and outlines the unexpected logic of inhibiting SGLT2 in the diabetic kidney.
Collapse
Affiliation(s)
- Ban Liu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuliang Wang
- Department of Immunology, Nanjing Medical University, Nanjing, China
| | - Yangyang Zhang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Tongji University School of Medicine, Shanghai, China.,Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Biao Yan
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China.,Eye Institute, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
29
|
Varsamis P, Formosa MF, Larsen RN, Reddy-Luthmoodoo M, Jennings GL, Cohen ND, Grace M, Hawley JA, Devlin BL, Owen N, Dunstan DW, Dempsey PC, Kingwell BA. Between-meal sucrose-sweetened beverage consumption impairs glycaemia and lipid metabolism during prolonged sitting: A randomized controlled trial. Clin Nutr 2019; 38:1536-1543. [PMID: 30217471 DOI: 10.1016/j.clnu.2018.08.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 08/08/2018] [Accepted: 08/20/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Chronic overconsumption of sugar-sweetened beverages (SSBs) is associated with unfavourable health effects, including promotion of obesity. However, the acute effects of consuming SSBs on glucose and lipid metabolism remain to be characterized in a real-world, post-prandial context of prolonged sitting. We quantified the acute effects of between-meal SSB consumption compared with water, on glucose and lipid metabolism in habitual soft drink consumers during prolonged sitting. METHODS Twenty-eight overweight or obese young adults [15 males; 23 ± 3 (mean ± SD) years, body mass index (BMI) 31.0 ± 3.6 kg/m2) participated. During uninterrupted sitting and following standardized breakfast and lunch meals, each participant completed two 7-h conditions on separate days in a randomized, crossover design study. For each condition, participants consumed either a sucrose SSB or water mid-morning and mid-afternoon. Peak responses and total area under the curve (tAUC) over 7 h for blood glucose, insulin, C-peptide, triglyceride and non-esterified fatty acid (NEFA) concentrations were quantified and compared. RESULTS Compared to water, SSB consumption significantly increased the peak responses for blood glucose (20 ± 4% (mean ± SEM)), insulin (43 ± 15%) and C-peptide (21 ± 6%) concentrations. The tAUC for all these parameters was also increased by SSB consumption. The tAUC for triglycerides was 15 ± 5% lower after SSBs and this was driven by males (P < 0.05), as females showed no difference between conditions. The tAUC for NEFAs was 13 ± 5% lower after the SSB condition (P < 0.05). CONCLUSIONS Between-meal SSB consumption significantly elevated plasma glucose responses, associated with a sustained elevation in plasma insulin throughout a day of prolonged sitting. The SSB-induced reduction in circulating triglycerides and NEFAs indicates significant modulation of lipid metabolism, particularly in males. These metabolic effects may contribute to the development of metabolic disease when SSB consumption is habitual and co-occurring with prolonged sitting. Clinical Trial Registry number: ACTRN12616000840482, https://anzctr.org.au/Trial/Registration/TrialReview.aspx?ACTRN=12616000840482.
Collapse
Affiliation(s)
- Pia Varsamis
- Baker Heart & Diabetes Institute, Melbourne, Australia; Department of Physiology, Faculty of Medicine, Nursing & Health Sciences, Monash University, Melbourne, Australia.
| | | | | | | | - Garry L Jennings
- Baker Heart & Diabetes Institute, Melbourne, Australia; Sydney Medical School, University of Sydney, Sydney, Australia
| | - Neale D Cohen
- Baker Heart & Diabetes Institute, Melbourne, Australia
| | - Megan Grace
- Baker Heart & Diabetes Institute, Melbourne, Australia; Department of Physiology, Faculty of Medicine, Nursing & Health Sciences, Monash University, Melbourne, Australia
| | - John A Hawley
- Exercise & Nutrition Research Programme, Mary MacKillop Institute of Health Research, Australian Catholic University, Melbourne, Victoria, Australia
| | - Brooke L Devlin
- Exercise & Nutrition Research Programme, Mary MacKillop Institute of Health Research, Australian Catholic University, Melbourne, Victoria, Australia
| | - Neville Owen
- Baker Heart & Diabetes Institute, Melbourne, Australia; Central Clinical School, Faculty of Medicine, Nursing & Health Sciences, Monash University, Melbourne, Australia; School of Sport Science, Exercise and Health, The University of Western Australia, Perth, Australia; Swinburne University of Technology, Melbourne, Australia; School of Population and Global Health, The University of Melbourne, Melbourne, Australia
| | - David W Dunstan
- Baker Heart & Diabetes Institute, Melbourne, Australia; Central Clinical School, Faculty of Medicine, Nursing & Health Sciences, Monash University, Melbourne, Australia; Centre of Physical Activity and Nutrition Research, School of Exercise and Nutrition Sciences, Deakin University, Melbourne, Australia; Exercise & Nutrition Research Programme, Mary MacKillop Institute of Health Research, Australian Catholic University, Melbourne, Victoria, Australia; School of Public Health, The University of Queensland, Brisbane, Australia; School of Sport Science, Exercise and Health, The University of Western Australia, Perth, Australia
| | - Paddy C Dempsey
- Baker Heart & Diabetes Institute, Melbourne, Australia; Swinburne University of Technology, Melbourne, Australia
| | - Bronwyn A Kingwell
- Baker Heart & Diabetes Institute, Melbourne, Australia; Department of Physiology, Faculty of Medicine, Nursing & Health Sciences, Monash University, Melbourne, Australia; Central Clinical School, Faculty of Medicine, Nursing & Health Sciences, Monash University, Melbourne, Australia
| |
Collapse
|
30
|
Liu W, Mao Y, Schoenborn J, Wang Z, Tang G, Tang X. Whole blueberry protects pancreatic beta-cells in diet-induced obese mouse. Nutr Metab (Lond) 2019; 16:34. [PMID: 31139236 PMCID: PMC6530052 DOI: 10.1186/s12986-019-0363-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 05/06/2019] [Indexed: 12/20/2022] Open
Abstract
Background Blueberry is rich in bioactive substances and possesses powerful antioxidant potential, which can protect against oxidant-induced and inflammatory cell damage and cytotoxicity. The aim of this study was to determine how blueberry affects glucose metabolism and pancreatic β-cell proliferation in high fat diet (HFD)-induced obese mice. Methods Wild type male mice at age of 4 weeks received two different kinds of diets: high-fat diet (HFD) containing 60% fat or modified HFD supplemented with 4% (wt:wt) freeze-dried whole blueberry powder (HFD + B) for 14 weeks. A separate experiment was performed in mice fed with low-fat diet (LFD) containing 10% fat or modified LFD + B supplemented with 4% (wt:wt) freeze-dried whole blueberry powder. The metabolic parameters including blood glucose and insulin levels, glucose and insulin tolerances were measured. Results Blueberry-supplemented diet significantly increased insulin sensitivity and glucose tolerance in HFD + B mice compared to HFD mice. However, no difference was observed in blood glucose and insulin sensitivity between LFD + B and LFD mice. In addition, blueberry increased β-cell survival and prevented HFD-induced β-cell expansion. The most important finding was the observation of presence of small scattered islets in blueberry treated obese mice, which may reflect a potential role of blueberry in regenerating pancreatic β-cells. Conclusions Blueberry-supplemented diet can prevent obesity-induced insulin resistance by improving insulin sensitivity and protecting pancreatic β-cells. Blueberry supplementation has the potential to protect and improve health conditions for both type 1 and type 2 diabetes patients. Electronic supplementary material The online version of this article (10.1186/s12986-019-0363-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Weixiang Liu
- Department of Biological Sciences, Michigan Technological University, 1400 Townsend Dr, Houghton, MI 49931 USA
| | - Yiping Mao
- Department of Biological Sciences, Michigan Technological University, 1400 Townsend Dr, Houghton, MI 49931 USA
| | - Jacob Schoenborn
- Department of Biological Sciences, Michigan Technological University, 1400 Townsend Dr, Houghton, MI 49931 USA
| | - Zhihong Wang
- Department of Biological Sciences, Michigan Technological University, 1400 Townsend Dr, Houghton, MI 49931 USA
| | - Guiliang Tang
- Department of Biological Sciences, Michigan Technological University, 1400 Townsend Dr, Houghton, MI 49931 USA
| | - Xiaoqing Tang
- Department of Biological Sciences, Michigan Technological University, 1400 Townsend Dr, Houghton, MI 49931 USA
| |
Collapse
|
31
|
Terenzi DC, Trac JZ, Teoh H, Gerstein HC, Bhatt DL, Al-Omran M, Verma S, Hess DA. Vascular Regenerative Cell Exhaustion in Diabetes: Translational Opportunities to Mitigate Cardiometabolic Risk. Trends Mol Med 2019; 25:640-655. [PMID: 31053416 DOI: 10.1016/j.molmed.2019.03.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/25/2019] [Accepted: 03/28/2019] [Indexed: 12/17/2022]
Abstract
Ischemic cardiovascular complications remain a major cause of mortality in people with type 2 diabetes (T2D). Individuals with T2D may have a reduced ability to revascularize ischemic tissues due to abnormal production of circulating provascular progenitor cells. This 'regenerative cell exhaustion' process is intensified by increasing oxidative stress and inflammation and during T2D progression. Chronic exhaustion may be mediated by changes in the bone marrow microenvironment that dysregulate the wingless related integration site network, a central pathway maintaining the progenitor cell pool. Restoration of vascular regenerative cell production by reducing glucotoxicity with contemporary antihyperglycemic agents, by reducing systemic inflammation postbariatric surgery, or by modulating progenitor cell provascular functions using exosomal manipulation, may provide unique approaches for mitigating ischemic disease.
Collapse
Affiliation(s)
- Daniella C Terenzi
- Division of Cardiac Surgery, St. Michael's Hospital, Toronto, ON, M5 B 1W8, Canada; Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, M5 B 1T8, Canada; Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, M5 B 1T8, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, M5S 1A1, Canada
| | - Justin Z Trac
- Division of Cardiac Surgery, St. Michael's Hospital, Toronto, ON, M5 B 1W8, Canada; Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, M5 B 1T8, Canada; Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, M5 B 1T8, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Hwee Teoh
- Division of Cardiac Surgery, St. Michael's Hospital, Toronto, ON, M5 B 1W8, Canada; Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, M5 B 1T8, Canada; Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, M5 B 1T8, Canada; Division of Endocrinology and Metabolism, St. Michael's Hospital Medical Centre, Toronto, ON, M5C 2T2, Canada
| | - Hertzel C Gerstein
- Division of Endocrinology and Metabolism, McMaster University and Hamilton Health Sciences, Population Health Research Institute, Hamilton, ON, L8S 4K1, Canada
| | - Deepak L Bhatt
- Brigham and Women's Hospital, Heart and Vascular Center, Harvard Medical School, Boston, MA 02115, USA
| | - Mohammed Al-Omran
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, M5 B 1T8, Canada; Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, M5 B 1T8, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, M5S 1A1, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, M5S 1A8, Canada; Division of Vascular Surgery, St. Michael's Hospital, Toronto, ON, M5 B 1W8, Canada; Department of Surgery, University of Toronto, Toronto, ON, M5T 1P5, Canada; Department of Surgery, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Subodh Verma
- Division of Cardiac Surgery, St. Michael's Hospital, Toronto, ON, M5 B 1W8, Canada; Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, M5 B 1T8, Canada; Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, M5 B 1T8, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, M5S 1A1, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, M5S 1A8, Canada; Department of Surgery, University of Toronto, Toronto, ON, M5T 1P5, Canada
| | - David A Hess
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, M5S 1A8, Canada; Division of Vascular Surgery, St. Michael's Hospital, Toronto, ON, M5 B 1W8, Canada; Department of Physiology and Pharmacology, Western University, London, ON, N6A 5C1, Canada; Robarts Research Institute, Western University, London, ON, N6A 5B7, Canada.
| |
Collapse
|
32
|
A carbohydrate-reduced high-protein diet acutely decreases postprandial and diurnal glucose excursions in type 2 diabetes patients. Br J Nutr 2019; 119:910-917. [PMID: 29644957 DOI: 10.1017/s0007114518000521] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The aim of the study was to assess whether a simple substitution of carbohydrate in the conventionally recommended diet with protein and fat would result in a clinically meaningful reduction in postprandial hyperglycaemia in subjects with type 2 diabetes mellitus (T2DM). In all, sixteen subjects with T2DM treated with metformin only, fourteen male, with a median age of 65 (43-70) years, HbA1c of 6·5 % (47 mmol/l) (5·5-8·3 % (37-67 mmol/l)) and a BMI of 30 (sd 4·4) kg/m2 participated in the randomised, cross-over study. A carbohydrate-reduced high-protein (CRHP) diet was compared with an iso-energetic conventional diabetes (CD) diet. Macronutrient contents of the CRHP/CD diets consisted of 31/54 % energy from carbohydrate, 29/16 % energy from protein and 40/30 % energy from fat, respectively. Each diet was consumed on 2 consecutive days in a randomised order. Postprandial glycaemia, pancreatic and gut hormones, as well as satiety, were evaluated at breakfast and lunch. Compared with the CD diet, the CRHP diet reduced postprandial AUC of glucose by 14 %, insulin by 22 % and glucose-dependent insulinotropic polypeptide by 17 % (all P<0·001), respectively. Correspondingly, glucagon AUC increased by 33 % (P<0·001), cholecystokinin by 24 % (P=0·004) and satiety scores by 7 % (P=0·035), respectively. A moderate reduction in carbohydrate with an increase in fat and protein in the diet, compared with an energy-matched CD diet, greatly reduced postprandial glucose excursions and resulted in increased satiety in patients with well-controlled T2DM.
Collapse
|
33
|
Aktay G, Gürsoy ŞÖ, Uyumlu U, Ünüvar S, İlhan N. Protective effect of atorvastatin on oxidative stress in streptozotocin-induced diabetic rats independently their lipid-lowering effects. J Biochem Mol Toxicol 2019; 33:e22295. [PMID: 30657622 DOI: 10.1002/jbt.22295] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 11/01/2018] [Accepted: 01/03/2019] [Indexed: 12/19/2022]
Abstract
In the present study, we investigate the effects of atorvastatin on the lipid profile, oxidative stress, and liver enzyme markers, and its protective activity against diabetic complications, in streptozotocin (STZ)-induced diabetic rats. Fasting blood glucose (FBG), triglyceride (TG), total cholesterol (TC), and high-density lipoprotein (HDL) levels, as well as alanine aminotransferase (ALT) and aspartate aminotransferase (AST) enzyme activities, were measured 7 weeks after the administration of STZ and atorvastatin. Thiobarbituric acid reactive substances (TBARS), non-protein associated sulfhydryl (NP-SH), total sulfhydryl (T-SH), and nitric oxide (NO) levels were measured to evaluate oxidative stress. Atorvastatin was found to inhibit ALT and AST activities and to reduce FBG levels in rats with STZ-induced diabetes. Moreover, atorvastatin treatment significantly reduced lipid peroxidation in kidney, heart, and eye tissues (P < 0.001, for all), and resulted in a significant increase in NP-SH levels in brain tissues (P < 0.001). Total NO and nitrate levels increased significantly after atorvastatin treatment (P < 0.01). Our results revealed that atorvastatin has a protective effect against STZ-induced oxidative damage by reducing TBARS levels and increasing NP-SH levels, has a hepatoprotective effect by decreasing ALT and AST activities. It also shows the antihyperglycemic activity by lowering FBG levels.
Collapse
Affiliation(s)
- Göknur Aktay
- Department of Pharmacology, Faculty of Pharmacy, İnönü University, Malatya, Turkey
| | - Şule Öner Gürsoy
- Department of Pharmacology, Faculty of Pharmacy, İnönü University, Malatya, Turkey
| | - Umut Uyumlu
- Department of Pharmacology, Faculty of Pharmacy, İnönü University, Malatya, Turkey
| | - Songül Ünüvar
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, İnönü University, Malatya, Turkey
| | - Nevin İlhan
- Department of Biochemistry, Faculty of Medicine, Fırat University, Elazığ, Turkey
| |
Collapse
|
34
|
Graus-Nunes F, Souza-Mello V. The renin-angiotensin system as a target to solve the riddle of endocrine pancreas homeostasis. Biomed Pharmacother 2018; 109:639-645. [PMID: 30404071 DOI: 10.1016/j.biopha.2018.10.191] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 10/30/2018] [Accepted: 10/31/2018] [Indexed: 10/27/2022] Open
Abstract
Local renin-angiotensin system (RAS) in the pancreas is linked to the modulation of glucose-stimulated insulin secretion (GSIS) in beta cells and insulin sensitivity in target tissues, emerging as a promising tool in the prevention and/or treatment of obesity, diabetes, and systemic arterial hypertension. Insulin resistance alters pancreatic islet cell distribution and morphology and hypertrophied islets exhibit upregulated angiotensin II type 1 receptor, which drives oxidative stress, apoptosis, and fibrosis, configuring beta cell dysfunction and diminishing islet lifespan. Pharmacological modulation of RAS has shown beneficial effects in diet-induced obesity model, mainly related to the translational potential that angiotensin receptor blockers and ECA2/ANG (1-7)/MAS receptor axis modulation have when it comes to islet preservation and type 2 diabetes prevention and/or treatment. This review describes the existing evidence for different approaches to blocking RAS elements in the management of insulin resistance and diabetes and focuses on islet remodeling and GSIS in rodents and humans.
Collapse
Affiliation(s)
- Francielle Graus-Nunes
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Brazil
| | - Vanessa Souza-Mello
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Brazil.
| |
Collapse
|
35
|
Rachdaoui N, Polo-Parada L, Ismail-Beigi F. Prolonged Exposure to Insulin Inactivates Akt and Erk 1/2 and Increases Pancreatic Islet and INS1E β-Cell Apoptosis. J Endocr Soc 2018; 3:69-90. [PMID: 30697602 PMCID: PMC6344346 DOI: 10.1210/js.2018-00140] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 10/29/2018] [Indexed: 12/18/2022] Open
Abstract
Chronic hyperinsulinemia, in vivo, increases the resistance of peripheral tissues to insulin by desensitizing insulin signaling. Insulin, in a heterologous manner, can also cause IGF-1 resistance. The aim of the current study was to investigate whether insulin-mediated insulin and IGF-1 resistance develops in pancreatic β-cells and whether this resistance results in β-cell decompensation. Chronic exposure of rat islets or INS1E β-cells to increasing concentrations of insulin decreased AktS473 phosphorylation in response to subsequent acute stimulation with 10 nM insulin or IGF-1. Prolonged exposure to high insulin levels not only inhibited AktS473 phosphorylation, but it also resulted in a significant inhibition of the phosphorylation of P70S6 kinase and Erk1/2 phosphorylation in response to the acute stimulation by glucose, insulin, or IGF-1. Decreased activation of Akt, P70S6K, and Erk1/2 was associated with decreased insulin receptor substrate 2 tyrosine phosphorylation and insulin receptor β-subunit abundance; neither IGF receptor β-subunit content nor its phosphorylation were affected. These signaling impairments were associated with decreased SERCA2 expression, perturbed plasma membrane calcium current and intracellular calcium handling, increased endoplasmic reticulum stress markers such as eIF2αS51 phosphorylation and Bip (GRP78) expression, and increased islet and β-cell apoptosis. We demonstrate that prolonged exposure to high insulin levels induces not only insulin resistance, but in a heterologous manner causes resistance to IGF-1 in rat islets and insulinoma cells resulting in decreased cell survival. These findings suggest the possibility that chronic exposure to hyperinsulinemia may negatively affect β-cell mass by increasing β-cell apoptosis.
Collapse
Affiliation(s)
- Nadia Rachdaoui
- Division of Clinical and Molecular Endocrinology, Department of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Luis Polo-Parada
- Department of Medical Pharmacology and Physiology, Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
| | - Faramarz Ismail-Beigi
- Division of Clinical and Molecular Endocrinology, Department of Medicine, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
36
|
Samkani A, Skytte MJ, Thomsen MN, Astrup A, Deacon CF, Holst JJ, Madsbad S, Rehfeld JF, Krarup T, Haugaard SB. Acute Effects of Dietary Carbohydrate Restriction on Glycemia, Lipemia and Appetite Regulating Hormones in Normal-Weight to Obese Subjects. Nutrients 2018; 10:nu10091285. [PMID: 30213037 PMCID: PMC6163561 DOI: 10.3390/nu10091285] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 09/07/2018] [Accepted: 09/10/2018] [Indexed: 12/26/2022] Open
Abstract
Postprandial responses to food are highly dependent on the macronutrient composition of the diet. We investigated the acute effects of transition from the recommended moderately high carbohydrate (HC) diet towards a carbohydrate-reduced high-protein (CRHP) diet on postprandial glycemia, insulinemia, lipemia, and appetite-regulating hormones in non-diabetic adults. Fourteen subjects, including five males (Mean ± SD: age 62 ± 6.5; BMI 32 ± 7.6 kg/m2; hemoglobin A1c (HbA1c) 40 ± 3.0 mmol/mol; HOMA2-IR 2.1 ± 0.9) were included in this randomized, cross-over study. Iso-caloric diets were consumed for two consecutive days with a median wash-out period of 21 days (range 2–8 weeks) between diets (macronutrient energy composition: CRHP/HC; 31%/54% carbohydrate, 29%/16% protein, 40%/30% fat). Postprandial glucose, insulin secretion rate (ISR), triglycerides (TGs), non-esterified fatty acids (NEFAs), and satiety ratings were assessed after ingestion of breakfast (Br) and lunch (Lu), and gut hormones and glucagon were assessed after ingestion of Br. Compared with the HC diet, the CRHP diet reduced peak glucose concentrations (Br 11%, p = 0.024; Lu 11%, p < 0.001), glucose excursions (Br 80%, p = 0.20; Lu 85%, p < 0.001), and ISR (Br 31%; Lu 64%, both p < 0.001) whereas CRHP, as compared with HC, increased glucagon-like peptide-1 (Br 27%, p = 0.015) and glucagon values (Br 249%, p < 0.001). NEFA and TG levels increased in the CRHP diet as compared with the HC diet after Br, but no difference was found after Lu (NEFA Br 22%, p < 0.01; TG Br 42%, p = 0.012). Beta-cell glucose sensitivity, insulin clearance, cholecystokinin values, and subjective satiety ratings were unaffected. It is possible to achieve a reduction in postprandial glycemia and insulin without a deleterious effect on beta-cell glucose sensitivity by substituting part of dietary carbohydrate with iso-caloric protein and fat in subjects without type 2 diabetes mellitus (T2DM). The metabolic effects are more pronounced after the second meal.
Collapse
Affiliation(s)
- Amirsalar Samkani
- Department of Endocrinology, Copenhagen University Hospital, Bispebjerg, DK-2400 Copenhagen NV, Denmark.
| | - Mads J Skytte
- Department of Endocrinology, Copenhagen University Hospital, Bispebjerg, DK-2400 Copenhagen NV, Denmark.
| | - Mads N Thomsen
- Department of Endocrinology, Copenhagen University Hospital, Bispebjerg, DK-2400 Copenhagen NV, Denmark.
| | - Arne Astrup
- Department of Nutrition, Exercise and Sports, University of Copenhagen, DK-1017 Copenhagen K, Denmark.
| | - Carolyn F Deacon
- Endocrinology Research Section, Department of Biomedical Sciences, University of Copenhagen, DK-1017 Copenhagen K, Denmark.
- Section for Translational Physiology, NNF Center for Basic Metabolic Research, University of Copenhagen, DK-1017 Copenhagen K, Denmark.
| | - Jens J Holst
- Endocrinology Research Section, Department of Biomedical Sciences, University of Copenhagen, DK-1017 Copenhagen K, Denmark.
- Section for Translational Physiology, NNF Center for Basic Metabolic Research, University of Copenhagen, DK-1017 Copenhagen K, Denmark.
| | - Sten Madsbad
- Department of Endocrinology, Copenhagen University Hospital, Amager Hvidovre, DK-2650 Hvidovre, Denmark.
| | - Jens F Rehfeld
- Department of Clinical Biochemistry, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark.
| | - Thure Krarup
- Department of Endocrinology, Copenhagen University Hospital, Bispebjerg, DK-2400 Copenhagen NV, Denmark.
| | - Steen B Haugaard
- Department of Endocrinology, Copenhagen University Hospital, Bispebjerg, DK-2400 Copenhagen NV, Denmark.
- Department of Internal Medicine, Copenhagen University Hospital, Amager Hvidovre, DK-2650 Hvidovre, Denmark.
| |
Collapse
|
37
|
Gómez-Guzmán M, Rodríguez-Nogales A, Algieri F, Gálvez J. Potential Role of Seaweed Polyphenols in Cardiovascular-Associated Disorders. Mar Drugs 2018; 16:E250. [PMID: 30060542 PMCID: PMC6117645 DOI: 10.3390/md16080250] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 07/22/2018] [Accepted: 07/26/2018] [Indexed: 01/09/2023] Open
Abstract
The beneficial effects of various polyphenols with plant origins on different cardiovascular-associated disorders, such as hypertension, diabetes mellitus type 2 and metabolic syndrome are well known. Recently, marine crude-drugs are emerging as potential treatments in many noncommunicable conditions, including those involving the cardiovascular system. Among the active compounds responsible for these activities, seaweed polyphenols seem to play a key role. The aim of the present review is to summarise the current knowledge about the beneficial effects reported for edible seaweed polyphenols in the amelioration of these prevalent conditions, focusing on both preclinical and clinical studies. This review will help to establish the basis for future studies in this promising field.
Collapse
Affiliation(s)
- Manuel Gómez-Guzmán
- Department of Pharmacology, School of Pharmacy, University of Granada, 18071 Granada, Spain.
- Instituto de Investigación Biosanitaria de Granada (Ibs.GRANADA), 18071 Granada, Spain.
| | - Alba Rodríguez-Nogales
- Instituto de Investigación Biosanitaria de Granada (Ibs.GRANADA), 18071 Granada, Spain.
- CIBER-EHD, Department of Pharmacology, Centre for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain.
| | - Francesca Algieri
- Instituto de Investigación Biosanitaria de Granada (Ibs.GRANADA), 18071 Granada, Spain.
- CIBER-EHD, Department of Pharmacology, Centre for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain.
| | - Julio Gálvez
- Instituto de Investigación Biosanitaria de Granada (Ibs.GRANADA), 18071 Granada, Spain.
- CIBER-EHD, Department of Pharmacology, Centre for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain.
| |
Collapse
|
38
|
Lee HA, Lee JH, Han JS. 2,7"-Phloroglucinol-6,6'-bieckol protects INS-1 cells against high glucose-induced apoptosis. Biomed Pharmacother 2018; 103:1473-1481. [DOI: 10.1016/j.biopha.2018.04.129] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 04/17/2018] [Accepted: 04/17/2018] [Indexed: 10/17/2022] Open
|
39
|
Dempsey PC, Larsen RN, Winkler EAH, Owen N, Kingwell BA, Dunstan DW. Prolonged uninterrupted sitting elevates postprandial hyperglycaemia proportional to degree of insulin resistance. Diabetes Obes Metab 2018; 20:1526-1530. [PMID: 29431272 DOI: 10.1111/dom.13254] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 01/30/2018] [Accepted: 02/06/2018] [Indexed: 02/02/2023]
Abstract
Prolonged uninterrupted sitting is related adversely to cardiometabolic risk markers and postprandial hyperglycaemia, relative to sitting interrupted by regular brief activity breaks. However, whether the magnitude of hyperglycaemic responses to prolonged sitting is dependent upon the underlying degree of insulin resistance remains unclear. Data were pooled from 3 randomized cross-over laboratory-based trials (n = 62) that examined the postprandial blood glucose- and insulin-lowering effects of prolonged sitting vs sitting interrupted by regular brief activity breaks in overweight/obese adults who had normal or impaired glucose metabolism (2 trials) or type 2 diabetes not treated by insulin (1 trial). Corrected for study effects, the magnitude of differences in postprandial glucose and insulin responses between the 2 conditions was significantly exacerbated with poorer baseline levels of fasting glucose, insulin and/or surrogate markers of β-cell function and insulin resistance. This suggests that those with higher underlying levels of insulin resistance may derive greater metabolic benefits from regularly interrupting prolonged sitting than their healthier counterparts. If these findings can be replicated, they may have implications for future targeting and optimization of physical activity/sedentary behaviour interventions in the prevention and management of type 2 diabetes.
Collapse
Affiliation(s)
- Paddy C Dempsey
- Baker Heart & Diabetes Institute, Melbourne, Australia
- Swinburne University of Technology, Melbourne, Australia
| | | | | | - Neville Owen
- Baker Heart & Diabetes Institute, Melbourne, Australia
- Swinburne University of Technology, Melbourne, Australia
- School of Public Health, The University of Queensland, Brisbane, Australia
- Central Clinical School/Department of Epidemiology and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
- Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia
| | - Bronwyn A Kingwell
- Baker Heart & Diabetes Institute, Melbourne, Australia
- Central Clinical School/Department of Epidemiology and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
| | - David W Dunstan
- Baker Heart & Diabetes Institute, Melbourne, Australia
- School of Public Health, The University of Queensland, Brisbane, Australia
- Central Clinical School/Department of Epidemiology and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
- Institute of Physical Activity and Nutrition Research, School of Exercise and Nutrition Sciences, Deakin University, Melbourne, Australia
- Mary MacKillop Institute of Health Research, Australian Catholic University, Melbourne, Australia
- School of Sport Science, Exercise and Health, The University of Western Australia, Perth, Australia
| |
Collapse
|
40
|
Yan L. Redox imbalance stress in diabetes mellitus: Role of the polyol pathway. Animal Model Exp Med 2018; 1:7-13. [PMID: 29863179 PMCID: PMC5975374 DOI: 10.1002/ame2.12001] [Citation(s) in RCA: 166] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 01/16/2018] [Indexed: 12/14/2022] Open
Abstract
In diabetes mellitus, the polyol pathway is highly active and consumes approximately 30% glucose in the body. This pathway contains 2 reactions catalyzed by aldose reductase (AR) and sorbitol dehydrogenase, respectively. AR reduces glucose to sorbitol at the expense of NADPH, while sorbitol dehydrogenase converts sorbitol to fructose at the expense of NAD+, leading to NADH production. Consumption of NADPH, accumulation of sorbitol, and generation of fructose and NADH have all been implicated in the pathogenesis of diabetes and its complications. In this review, the roles of this pathway in NADH/NAD+ redox imbalance stress and oxidative stress in diabetes are highlighted. A potential intervention using nicotinamide riboside to restore redox balance as an approach to fighting diabetes is also discussed.
Collapse
Affiliation(s)
- Liang‐jun Yan
- Department of Pharmaceutical SciencesUNT System College of PharmacyUniversity of North Texas Health Science CenterFort WorthTXUSA
| |
Collapse
|
41
|
Lafferty RA, Flatt PR, Irwin N. Emerging therapeutic potential for peptide YY for obesity-diabetes. Peptides 2018; 100:269-274. [PMID: 29412828 DOI: 10.1016/j.peptides.2017.11.005] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 11/06/2017] [Accepted: 11/06/2017] [Indexed: 12/25/2022]
Abstract
The vast majority of research to date on the gut hormone Peptide YY (PYY) has focused on appetite suppression and body weight regulation effects. These biological actions are believed to occur through interaction of PYY with hypothalamic Y2 receptors. However, more recent studies have added additional knowledge to understanding of the physiological, and potential therapeutic, roles of PYY beyond obesity alone. Thus, PYY has now been shown to impart improvements in pancreatic beta-cell survival and function, with obvious benefits for diabetes. This effect has been linked mainly to binding and activation of Y1 receptors by PYY, but more evidence is still required in this regard. Given the potential therapeutic promise of PYY-derived compounds, and complexity of receptor interactions, it is important to fully understand the complete biological action profile of PYY. Therefore, the current review aims to compile, evaluate and summarise current knowledge on PYY, with particular emphasis on obesity and diabetes treatment, and the importance of specific Y receptor interactions for this.
Collapse
Affiliation(s)
- Ryan A Lafferty
- SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, Northern Ireland, BT52 1SA, UK
| | - Peter R Flatt
- SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, Northern Ireland, BT52 1SA, UK
| | - Nigel Irwin
- SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, Northern Ireland, BT52 1SA, UK.
| |
Collapse
|
42
|
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
|
43
|
Ottosson-Laakso E, Krus U, Storm P, Prasad RB, Oskolkov N, Ahlqvist E, Fadista J, Hansson O, Groop L, Vikman P. Glucose-Induced Changes in Gene Expression in Human Pancreatic Islets: Causes or Consequences of Chronic Hyperglycemia. Diabetes 2017; 66:3013-3028. [PMID: 28882899 DOI: 10.2337/db17-0311] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Accepted: 08/30/2017] [Indexed: 11/13/2022]
Abstract
Dysregulation of gene expression in islets from patients with type 2 diabetes (T2D) might be causally involved in the development of hyperglycemia, or it could develop as a consequence of hyperglycemia (i.e., glucotoxicity). To separate the genes that could be causally involved in pathogenesis from those likely to be secondary to hyperglycemia, we exposed islets from human donors to normal or high glucose concentrations for 24 h and analyzed gene expression. We compared these findings with gene expression in islets from donors with normal glucose tolerance and hyperglycemia (including T2D). The genes whose expression changed in the same direction after short-term glucose exposure, as in T2D, were considered most likely to be a consequence of hyperglycemia. Genes whose expression changed in hyperglycemia but not after short-term glucose exposure, particularly those that also correlated with insulin secretion, were considered the strongest candidates for causal involvement in T2D. For example, ERO1LB, DOCK10, IGSF11, and PRR14L were downregulated in donors with hyperglycemia and correlated positively with insulin secretion, suggesting a protective role, whereas TMEM132C was upregulated in hyperglycemia and correlated negatively with insulin secretion, suggesting a potential pathogenic role. This study provides a catalog of gene expression changes in human pancreatic islets after exposure to glucose.
Collapse
Affiliation(s)
- Emilia Ottosson-Laakso
- Lund University Diabetes Centre, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Ulrika Krus
- Lund University Diabetes Centre, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Petter Storm
- Lund University Diabetes Centre, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Rashmi B Prasad
- Lund University Diabetes Centre, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Nikolay Oskolkov
- Lund University Diabetes Centre, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Emma Ahlqvist
- Lund University Diabetes Centre, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - João Fadista
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Ola Hansson
- Lund University Diabetes Centre, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Leif Groop
- Lund University Diabetes Centre, Department of Clinical Sciences, Lund University, Malmö, Sweden
- Finnish Institute of Molecular Medicine, University of Helsinki, Helsinki, Finland
| | - Petter Vikman
- Lund University Diabetes Centre, Department of Clinical Sciences, Lund University, Malmö, Sweden
| |
Collapse
|
44
|
Lee CH, Chu CS, Tsai HJ, Ke LY, Lee HC, Yeh JL, Chen CH, Wu BN. Xanthine-derived KMUP-1 reverses glucotoxicity-activated Kv channels through the cAMP/PKA signaling pathway in rat pancreatic β cells. Chem Biol Interact 2017; 279:171-176. [PMID: 29183753 DOI: 10.1016/j.cbi.2017.11.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 11/23/2017] [Indexed: 10/18/2022]
Abstract
Hyperglycemia-associated glucotoxicity induces β-cell dysfunction and a reduction in insulin secretion. Voltage-dependent K+ (Kv) channels in pancreatic β-cells play a key role in glucose-dependent insulin secretion. KMUP-1, a xanthine derivative, has been demonstrated to modulate Kv channel activity in smooth muscles; however, the role of KMUP-1 in glucotoxicity-activated Kv channels in pancreatic β-cells remains unclear. In this study we examined the mechanisms by which KMUP-1 could inhibit high glucose (25 mM) activated Kv currents (IKv) in pancreatic β-cells. Pancreatic β-cells were isolated from Wistar rats and IKv was monitored by perforated patch-clamp recording. The peak IKv in high glucose-treated β-cells was ∼1.4-fold greater than for normal glucose (5.6 mM). KMUP-1 (1, 10, 30 μM) prevented high glucose-stimulated IKv in a concentration-dependent manner. Reduction of high glucose-activated IKv was also found for protein kinase A (PKA) activator 8-Br-cAMP (100 μM). Additionally, KMUP-1 (30 μM) current inhibition was reversed by the PKA inhibitor H-89 (1 μM). Otherwise, pretreatment with the PKC activator or inhibitor had no effect on IKv in high glucose exposure. In conclusion, glucotoxicity-diminished insulin secretion was due to IKv activation. KMUP-1 attenuated high glucose-stimulated IKv via the PKA but not the PKC signaling pathway. This finding provides evidence that KMUP-1 might be a promising agent for treating hyperglycemia-induced insulin resistance.
Collapse
Affiliation(s)
- Chien-Hsing Lee
- Department of Pharmacology, Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chih-Sheng Chu
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Han-Jie Tsai
- Department of Pharmacology, Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Liang-Yin Ke
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Hsiang-Chun Lee
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Jwu-Lai Yeh
- Department of Pharmacology, Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Chu-Huang Chen
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Vascular and Medicinal Research, Texas Heart Institute, Houston, TX 77030, USA
| | - Bin-Nan Wu
- Department of Pharmacology, Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
| |
Collapse
|
45
|
Kowalchuk C, Teo C, Wilson V, Chintoh A, Lam L, Agarwal SM, Giacca A, Remington GJ, Hahn MK. In male rats, the ability of central insulin to suppress glucose production is impaired by olanzapine, whereas glucose uptake is left intact. J Psychiatry Neurosci 2017; 42. [PMID: 29083297 PMCID: PMC5662464 DOI: 10.1503/jpn.170092] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Insulin receptors are widely expressed in the brain and may represent a crossroad between metabolic and cognitive disorders. Although antipsychotics, such as olanzapine, are the cornerstone treatment for schizophrenia, they are associated with high rates of type 2 diabetes and lack efficacy for illness-related cognitive deficits. Historically, this risk of diabetes was attributed to the weight gain propensity of antipsychotics, but recent work suggests antipsychotics can have weight-independent diabetogenic effects involving unknown brain-mediated mechanisms. Here, we examined whether antipsychotics disrupt central insulin action, hypothesizing that olanzapine would impair the well-established ability of central insulin to supress hepatic glucose production. METHODS Pancreatic euglycemic clamps were used to measure glucose kinetics alongside a central infusion of insulin or vehicle into the third ventricle. Male rats were pretreated with olanzapine or vehicle per our established model of acute olanzapine-induced peripheral insulin resistance. Groups included (central-peripheral) vehicle-vehicle (n = 11), insulin-vehicle (n = 10), insulin-olanzapine (n = 10) and vehicle-olanzapine (n = 8). RESULTS There were no differences in peripheral glucose or insulin levels. Unexpectedly, we showed that central insulin increased glucose uptake, and this effect was not perturbed by olanzapine. We replicated suppression of glucose production by insulin (clamp relative to basal: 77.9% ± 13.1%, all p < 0.05), an effect abolished by olanzapine (insulin-olanzapine: 7.7% ± 14%). LIMITATIONS This study used only male rats and an acute dose of olanzapine. CONCLUSION To our knowledge, this is the first study suggesting olanzapine may impair central insulin sensing, elucidating a potential mechanism of antipsychotic-induced diabetes and opening avenues of investigation related to domains of schizophrenia psychopathology.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Margaret K. Hahn
- Correspondence to: M.K. Hahn, Centre for Addiction and Mental Health, 250 College St, Toronto ON M5T 1R8;
| |
Collapse
|
46
|
The additive effects of atorvastatin and insulin on renal function and renal organic anion transporter 3 function in diabetic rats. Sci Rep 2017; 7:13532. [PMID: 29051569 PMCID: PMC5648883 DOI: 10.1038/s41598-017-13206-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 05/30/2017] [Indexed: 01/26/2023] Open
Abstract
Hyperglycemia-induced oxidative stress is usually found in diabetic condition. 3-hydroxy-3-methylglutaryl coenzyme-A (HMG-CoA) reductase inhibitors, statins, are widely used as cholesterol-lowering medication with several "pleiotropic" effects in diabetic patients. This study aims to evaluate whether the protective effects of atorvastatin and insulin on renal function and renal organic anion transporter 3 (Oat3) function involve the modulation of oxidative stress and pancreatic function in type 1 diabetic rats. Type 1 diabetes was induced by intraperitoneal injection of streptozotocin (50 mg/kg BW). Atorvastatin and insulin as single or combined treatment were given for 4 weeks after diabetic condition had been confirmed. Diabetic rats demonstrated renal function and renal Oat3 function impairment with an increased MDA level and decreased SOD protein expression concomitant with stimulation of renal Nrf2 and HO-1 protein expression. Insulin plus atorvastatin (combined) treatment effectively restored renal function as well as renal Oat3 function which correlated with the decrease in hyperglycemia and oxidative stress. Moreover, pancreatic inflammation and apoptosis in diabetic rats were ameliorated by the combined drugs treatment. Therefore, atorvastatin plus insulin seems to exert the additive effect in improving renal functionby alleviating hyperglycemiaand the modulation of oxidative stress, inflammation and apoptosis.
Collapse
|
47
|
Sugiyama Y, Kameshita I. Multi-PK antibodies: Powerful analytical tools to explore the protein kinase world. Biochem Biophys Rep 2017; 11:40-45. [PMID: 28955766 PMCID: PMC5614692 DOI: 10.1016/j.bbrep.2017.06.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 06/07/2017] [Accepted: 06/20/2017] [Indexed: 11/25/2022] Open
Abstract
Diverse biological events are regulated through protein phosphorylation mediated by protein kinases. Some of these protein kinases are known to be involved in the pathogenesis of various diseases. Although 518 protein kinase genes were identified in the human genome, it remains unclear how many and what kind of protein kinases are expressed and activated in cells and tissues under varying situations. To investigate cellular signaling by protein kinases, we developed monoclonal antibodies, designated as Multi-PK antibodies, that can recognize multiple protein kinases in various biological species. These Multi-PK antibodies can be used to profile the kinases expressed in cells and tissues, identify the kinases of special interest, and analyze protein kinase expression and phosphorylation state. Here we introduce some applications of Multi-PK antibodies to identify and characterize the protein kinases involved in epigenetics, glucotoxicity in type 2 diabetes, and pathogenesis of ulcerative colitis. In this review, we focus on the recently developed technologies for kinomics studies using the powerful analytical tools of Multi-PK antibodies. Multi-PK antibodies recognize a wide variety of protein kinases. New analytical methods using Multi-PK antibodies for protein kinase studies are explained. Kinomics studies using Multi-PK antibodies are introduced.
Collapse
Key Words
- 2D-PAGE, two-dimensional polyacrylamide gel electrophoresis
- CDKL5, cyclin-dependent kinase-like 5
- CNBr, cyanogen bromide
- CaMK, Ca2+/calmodulin-dependent protein kinase
- DCLK, double-cortin like protein kinase
- Dnmt1, DNA methyltransferase 1
- FAK, focal adhesion kinase
- IEF, isoelectric focusing
- IPG, immobilized pH gradient
- Kinomics
- MAPK, mitogen-activated protein kinase
- MeCP2, methylated-CpG-binding protein 2
- Monoclonal antibody
- Protein kinase
- Protein phosphorylation
- Proteomics
Collapse
|
48
|
Kain V, Halade GV. Metabolic and Biochemical Stressors in Diabetic Cardiomyopathy. Front Cardiovasc Med 2017; 4:31. [PMID: 28620607 PMCID: PMC5449449 DOI: 10.3389/fcvm.2017.00031] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 04/28/2017] [Indexed: 12/18/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) or diabetes-induced cardiac dysfunction is a direct consequence of uncontrolled metabolic syndrome and is widespread in US population and worldwide. Despite of the heterogeneous and distinct features of DCM, the clinical relevance of DCM is now becoming established. DCM progresses to pathological cardiac remodeling with the higher risk of heart attack and subsequent heart failure in diabetic patients. In this review, we emphasize lipid substrate quality and the phenotypic, metabolic, and biochemical stressors of DCM in the rodent and human pathophysiology. We discuss lipoxygenase signaling in the inflammatory pathway with multiple contributing and confounding factors leading to DCM. Additionally, emerging biochemical pathways are emphasized to make progress toward therapeutic advancement to treat DCM.
Collapse
Affiliation(s)
- Vasundhara Kain
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Ganesh V Halade
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
49
|
Liu C, Hao Y, Yin F, Zhang Y, Liu J. Geniposide protects pancreatic β cells from high glucose-mediated injury by activation of AMP-activated protein kinase. Cell Biol Int 2017; 41:544-554. [PMID: 28244615 DOI: 10.1002/cbin.10758] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 02/24/2017] [Indexed: 12/11/2022]
Abstract
Our previous works indicated that geniposide could regulate glucose-stimulated insulin secretion (GSIS), and improved chronic high glucose-induced dysfunctions in pancreatic β cells, but the molecular mechanisms remain largely unknown. In the present study, we investigated the role of 5'-AMP-activated protein kinase (AMPK) in high glucose induced cell injury and explored the associated molecular mechanisms in rat INS-1 pancreatic β cells. Data suggested that geniposide obviously prevented the cell damage induced by high (25 mM) glucose in INS-1 cells, which increased the protein levels of cell apoptosis-associated enzymes, including heme oxygenase-1 (HO-1), and Bcl-2, but apparently attenuated the protein level of Bax, an apoptotic protein. In addition, Compound C, an AMPK inhibitor, remarkably inhibited the effects of geniposide on the protein levels of HO-1, Bcl-2, and Bax, but AICAR, an AMPK activator, potentiated the role of geniposide on the protein levels of HO-1, Bcl-2, and Bax. More importantly, geniposide directly prevented the cleavage of caspase-3 induced by high glucose, and this effect was also evidently prohibited by the pre-incubation of compound C in high glucose-treated INS-1 cells. Furthermore, using the method of RNA interfere, we further proved that treatment with AMPK siRNA attenuated the effects of geniposide on the apoptosis-associated proteins and cell viability. All these data suggest that AMPK plays a crucial role on geniposide antagonizing high glucose-induced pancreatic β cells injury.
Collapse
Affiliation(s)
- Chunyan Liu
- Chongqing Key Lab of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing, 400054, China
| | - Yanan Hao
- Chongqing Key Lab of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing, 400054, China
| | - Fei Yin
- Chongqing Key Lab of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing, 400054, China
| | - Yonglan Zhang
- Chongqing Key Lab of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing, 400054, China
| | - Jianhui Liu
- Chongqing Key Lab of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing, 400054, China
| |
Collapse
|
50
|
Hao T, Zhang H, Li S, Tian H. Glucagon-like peptide 1 receptor agonist ameliorates the insulin resistance function of islet β cells via the activation of PDX-1/JAK signaling transduction in C57/BL6 mice with high-fat diet-induced diabetes. Int J Mol Med 2017; 39:1029-1036. [PMID: 28290604 DOI: 10.3892/ijmm.2017.2910] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 02/21/2017] [Indexed: 11/05/2022] Open
Abstract
Long-term exposure to a high-fat diet (HFD) causes glucotoxicity and lipotoxicity in islet β cells and leads to the development of metabolic dysfunctions. Reductions in pancreatic and duodenal homeobox-1 (PDX-1) expression have been shown to induce type 2 diabetes mellitus by causing impairments to islet β cells. Glucagon-like peptide 1 (GLP-1) treatment reduces endogenous insulin resistance in HFD-induced type 2 diabetes mellitus. In the present study, the underlying mechanism by which GLP-1 exerts its function in type 2 diabetes mellitus was investigated. The effect of liraglutide (GLP-1 receptor agonist) administration on glucose tolerance, insulin release, and glucose-dependent insulinotropic polypeptide level was detected in a HFD-induced diabetes C57/BL6 mouse model. Moreover, the role of liraglutide administration on the activity of PDX-1 was quantified to demonstrate the association between the two indicators. The results showed that administration of liraglutide could ameliorate the impairments to β cells due to HFD consumption. Liraglutide restored the insulin capacity and stimulated glucose disposal by improving the function and increasing the number of islet β cells. Furthermore, the hyperplasia and redundant function of islet α cells were inhibited by liraglutide treatment as well. At the molecular level, administration of liraglutide induced the expression of PDX-1, MafA, p-JAK2 and p-Stat3 in HFD model to relatively normal levels. It was suggested that the effect of liraglutide-induced activation of GLP-1 was exerted via activation of PDX-1 rather than its function in decreasing body weight. The study demonstrated that GLP-1 played an essential role in type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Tao Hao
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Hongtao Zhang
- Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Sheyu Li
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Haoming Tian
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|