1
|
Kim JY, Lee J, Kim SG, Kim NH. Recent Glycemia Is a Major Determinant of β-Cell Function in Type 2 Diabetes Mellitus. Diabetes Metab J 2024; 48:1135-1146. [PMID: 38889769 PMCID: PMC11621653 DOI: 10.4093/dmj.2023.0359] [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: 10/11/2023] [Accepted: 03/26/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGRUOUND Progressive deterioration of β-cell function is a characteristic of type 2 diabetes mellitus (T2DM). We aimed to investigate the relative contributions of clinical factors to β-cell function in T2DM. METHODS In a T2DM cohort of 470 adults (disease duration 0 to 41 years), β-cell function was estimated using insulinogenic index (IGI), disposition index (DI), oral disposition index (DIO), and homeostasis model assessment of β-cell function (HOMA-B) derived from a 75 g oral glucose tolerance test (OGTT). The relative contributions of age, sex, disease duration, body mass index, glycosylated hemoglobin (HbA1c) levels (at the time of the OGTT), area under the curve of HbA1c over time (HbA1c AUC), coefficient of variation in HbA1c (HbA1c CV), and antidiabetic agents use were compared by standardized regression coefficients. Longitudinal analyses of these indices were also performed. RESULTS IGI, DI, DIO, and HOMA-B declined over time (P<0.001 for all). Notably, HbA1c was the most significant factor affecting IGI, DI, DIO, and HOMA-B in the multivariable regression analysis. Compared with HbA1c ≥9%, DI was 1.9-, 2.5-, 3.7-, and 5.5-fold higher in HbA1c of 8%-<9%, 7%-<8%, 6%-<7%, and <6%, respectively, after adjusting for confounding factors (P<0.001). Conversely, β-cell function was not affected by the type or duration of antidiabetic agents, HbA1c AUC, or HbA1c CV. The trajectories of the IGI, DI, DIO, and HOMA-B mirrored those of HbA1c. CONCLUSION β-Cell function declines over time; however, it is flexible, being largely affected by recent glycemia in T2DM.
Collapse
Affiliation(s)
- Ji Yoon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
- Division of Endocrinology and Metabolism, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jiyoon Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Sin Gon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Nam Hoon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
2
|
Lafferty RA, Flatt PR, Irwin N. NPYR modulation: Potential for the next major advance in obesity and type 2 diabetes management? Peptides 2024; 179:171256. [PMID: 38825012 DOI: 10.1016/j.peptides.2024.171256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/13/2024] [Accepted: 05/30/2024] [Indexed: 06/04/2024]
Abstract
The approval of the glucagon-like peptide 1 (GLP-1) mimetics semaglutide and liraglutide for management of obesity, independent of type 2 diabetes (T2DM), has initiated a resurgence of interest in gut-hormone derived peptide therapies for the management of metabolic diseases, but side-effect profile is a concern for these medicines. However, the recent approval of tirzepatide for obesity and T2DM, a glucose-dependent insulinotropic polypeptide (GIP), GLP-1 receptor co-agonist peptide therapy, may provide a somewhat more tolerable option. Despite this, an increasing number of non-incretin alternative peptides are in development for obesity, and it stands to reason that other hormones will take to the limelight in the coming years, such as peptides from the neuropeptide Y family. This narrative review outlines the therapeutic promise of the neuropeptide Y family of peptides, comprising of the 36 amino acid polypeptides neuropeptide Y (NPY), peptide tyrosine-tyrosine (PYY) and pancreatic polypeptide (PP), as well as their derivatives. This family of peptides exerts a number of metabolically relevant effects such as appetite regulation and can influence pancreatic beta-cell survival. Although some of these actions still require full translation to the human setting, potential therapeutic application in obesity and type 2 diabetes is conceivable. However, like GLP-1 and GIP, the endogenous NPY, PYY and PP peptide forms are subject to rapid in vivo degradation and inactivation by the serine peptidase, dipeptidyl-peptidase 4 (DPP-4), and hence require structural modification to prolong circulating half-life. Numerous protective modification strategies are discussed in this regard herein, alongside related impact on biological activity profile and therapeutic promise.
Collapse
Affiliation(s)
- Ryan A Lafferty
- Diabetes Research Centre, Ulster University, Coleraine, Northern Ireland BT52 1SA, UK.
| | - Peter R Flatt
- Diabetes Research Centre, Ulster University, Coleraine, Northern Ireland BT52 1SA, UK
| | - Nigel Irwin
- Diabetes Research Centre, Ulster University, Coleraine, Northern Ireland BT52 1SA, UK
| |
Collapse
|
3
|
Webster KL, Mirmira RG. Beta cell dedifferentiation in type 1 diabetes: sacrificing function for survival? Front Endocrinol (Lausanne) 2024; 15:1427723. [PMID: 38904049 PMCID: PMC11187278 DOI: 10.3389/fendo.2024.1427723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 05/27/2024] [Indexed: 06/22/2024] Open
Abstract
The pathogeneses of type 1 and type 2 diabetes involve the progressive loss of functional beta cell mass, primarily attributed to cellular demise and/or dedifferentiation. While the scientific community has devoted significant attention to unraveling beta cell dedifferentiation in type 2 diabetes, its significance in type 1 diabetes remains relatively unexplored. This perspective article critically analyzes the existing evidence for beta cell dedifferentiation in type 1 diabetes, emphasizing its potential to reduce beta cell autoimmunity. Drawing from recent advancements in both human studies and animal models, we present beta cell identity as a promising target for managing type 1 diabetes. We posit that a better understanding of the mechanisms of beta cell dedifferentiation in type 1 diabetes is key to pioneering interventions that balance beta cell function and immunogenicity.
Collapse
Affiliation(s)
| | - Raghavendra G. Mirmira
- Kovler Diabetes Center and the Department of Medicine, The University of Chicago, Chicago, IL, United States
| |
Collapse
|
4
|
Gomez-Muñoz L, Dominguez-Bendala J, Pastori RL, Vives-Pi M. Immunometabolic biomarkers for partial remission in type 1 diabetes mellitus. Trends Endocrinol Metab 2024; 35:151-163. [PMID: 37949732 DOI: 10.1016/j.tem.2023.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/13/2023] [Accepted: 10/13/2023] [Indexed: 11/12/2023]
Abstract
Shortly after diagnosis of type 1 diabetes mellitus (T1DM) and initiation of insulin therapy, many patients experience a transient partial remission (PR) phase, also known as the honeymoon phase. This phase presents a potential therapeutic opportunity due to its association with immunoregulatory and β cell-protective mechanisms. However, the lack of biomarkers makes its characterization difficult. In this review, we cover the current literature addressing the discovery of new predictive and monitoring biomarkers that contribute to the understanding of the metabolic, epigenetic, and immunological mechanisms underlying PR. We further discuss how these peripheral biomarkers reflect attempts to arrest β cell autoimmunity and how these can be applied in clinical practice.
Collapse
Affiliation(s)
- Laia Gomez-Muñoz
- Immunology Section, Germans Trias i Pujol Research Institute, Universitat Autònoma de Barcelona, 08916 Badalona, Spain
| | - Juan Dominguez-Bendala
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Ricardo L Pastori
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Marta Vives-Pi
- Immunology Section, Germans Trias i Pujol Research Institute, Universitat Autònoma de Barcelona, 08916 Badalona, Spain; Ahead Therapeutics SL, 08193, Bellaterra, Barcelona, Spain.
| |
Collapse
|
5
|
Knebel UE, Peleg S, Dai C, Cohen-Fultheim R, Jonsson S, Poznyak K, Israeli M, Zamashanski L, Glaser B, Levanon EY, Powers AC, Klochendler A, Dor Y. Disrupted RNA editing in beta cells mimics early-stage type 1 diabetes. Cell Metab 2024; 36:48-61.e6. [PMID: 38128529 PMCID: PMC10843671 DOI: 10.1016/j.cmet.2023.11.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 09/18/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023]
Abstract
A major hypothesis for the etiology of type 1 diabetes (T1D) postulates initiation by viral infection, leading to double-stranded RNA (dsRNA)-mediated interferon response and inflammation; however, a causal virus has not been identified. Here, we use a mouse model, corroborated with human islet data, to demonstrate that endogenous dsRNA in beta cells can lead to a diabetogenic immune response, thus identifying a virus-independent mechanism for T1D initiation. We found that disruption of the RNA editing enzyme adenosine deaminases acting on RNA (ADAR) in beta cells triggers a massive interferon response, islet inflammation, and beta cell failure and destruction, with features bearing striking similarity to early-stage human T1D. Glycolysis via calcium enhances the interferon response, suggesting an actionable vicious cycle of inflammation and increased beta cell workload.
Collapse
Affiliation(s)
- Udi Ehud Knebel
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel; Department of Military Medicine and "Tzameret", Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel, and Medical Corps, Israel Defense Forces, Israel
| | - Shani Peleg
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Chunhua Dai
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Roni Cohen-Fultheim
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel; Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan, Israel
| | - Sara Jonsson
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Karin Poznyak
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Maya Israeli
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Liza Zamashanski
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Benjamin Glaser
- Department of Endocrinology and Metabolism, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Erez Y Levanon
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Alvin C Powers
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; VA Tennessee Valley Healthcare System, Nashville, TN 37212, USA
| | - Agnes Klochendler
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Yuval Dor
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
6
|
Taheri F, Panahi N, Vahidi A, Asadi M, Amoli MM, Goharifar N. Role of EP4 factor in paediatric type 1 diabetes mellitus: a comprehensive review focusing on the honeymoon period. Pediatr Endocrinol Diabetes Metab 2024; 30:227-246. [PMID: 39963060 PMCID: PMC11809557 DOI: 10.5114/pedm.2024.146686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 09/15/2024] [Indexed: 02/20/2025]
Abstract
The partial clinical recovery phase (PCRP), or "honeymoon period", is a temporary and partial restoration of b-cell function in patients with type 1 diabetes mellitus (T1DM), in which the immune system attacks and destroys insulin-producing b-cells. The underlying causes of PCRP are not fully understood, but they are believed to involve a combination of genetic and environmental factors. Recent research has suggested a potential link between a specific allele of the prostaglandin receptor EP4 (PTGER4) and the modulation of remission in individuals with T1DM. This review aims to provide an overview of current scientific findings on the biological functions and role of the EP4 receptor in T1DM, with a particular focus on its involvement in the PCR phase. It provides a comprehensive understanding of the mechanisms underlying PCRP, which can lead to the development of more effective treatment strategies for preserving b-cell function and prolonging the PCRP. The identification of specific biomarkers associated with the PCRP and the EP4 receptor enables early identification of individuals at lower risk of long-term complications, facilitating targeted interventions and personalised treatment approaches.
Collapse
Affiliation(s)
- Forough Taheri
- Metabolic Disorders Research Centre, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Iran
| | - Nekoo Panahi
- Metabolic Disorders Research Centre, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Iran
- Endocrinology and Metabolism Research Centre, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Iran
| | - Aida Vahidi
- Metabolic Disorders Research Centre, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Iran
| | - Mojgan Asadi
- Metabolic Disorders Research Centre, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Iran
| | - Mahsa M. Amoli
- Metabolic Disorders Research Centre, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Iran
| | - Naieme Goharifar
- Department of Biology, Faculty of Science and Technology, ACECR Institute of Higher Education, Isfahan, Iran
| |
Collapse
|
7
|
Shapey IM, Summers A, Yiannoullou P, Fullwood C, Augustine T, Rutter MK, van Dellen D. Donor noradrenaline use is associated with better allograft survival in recipients of pancreas transplantation. Ann R Coll Surg Engl 2024; 106:19-28. [PMID: 36927080 PMCID: PMC10757882 DOI: 10.1308/rcsann.2022.0161] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2022] [Indexed: 03/18/2023] Open
Abstract
INTRODUCTION Outcomes following pancreas transplantation are suboptimal and better donor selection is required to improve this. Vasoactive drugs (VaD) are commonly used to correct the abnormal haemodynamics of organ donors in intensive care units. VaDs can differentially affect insulin secretion positively (dobutamine) or negatively (noradrenaline). The hypothesis was that some VaDs might induce beta-cell stress or rest and therefore impact pancreas transplant outcomes. The aim of the study was to assess relationships between VaD use and pancreas transplant graft survival. METHODS Data from the UK Transplant Registry on all pancreas transplants performed between 2004 and 2016 with complete follow-up data were included. Univariable- and multivariable-adjusted Cox regression analyses determined risks of graft failure associated with VaD use. RESULTS In 2,183 pancreas transplants, VaDs were used in the following numbers of donors: dobutamine 76 (3.5%), dopamine 84 (3.8%), adrenaline 161 (7.4%), noradrenaline 1,589 (72.8%) and vasopressin 1,219 (55.8%). In multivariable models, adjusted for covariates and the co-administration of other VaDs, noradrenaline use (vs non-use) was a strong predictor of better graft survival (hazard ratio [95% confidence interval] 0.77 [0.64-0.94], p = 0.01). CONCLUSIONS Noradrenaline use was associated with better graft survival in models adjusted for donor and recipient variables - this may be related to inhibition of pancreatic insulin secretion initiating pancreatic beta-cell 'rest'. Further research is required to replicate these findings and establish whether relationships are causal. Identification of alternative methods of inducing beta-cell rest could be valuable in improving graft outcomes.
Collapse
Affiliation(s)
- IM Shapey
- University of Manchester, UK
- Manchester University NHS Foundation Trust, UK
| | - A Summers
- Manchester University NHS Foundation Trust, UK
| | | | - C Fullwood
- University of Manchester, UK
- Manchester University NHS Foundation Trust, UK
| | - T Augustine
- Manchester University NHS Foundation Trust, UK
| | - MK Rutter
- University of Manchester, UK
- Manchester University NHS Foundation Trust, UK
| | | |
Collapse
|
8
|
Makki BE, Rahman S. Alzheimer's Disease in Diabetic Patients: A Lipidomic Prospect. Neuroscience 2023; 530:79-94. [PMID: 37652288 DOI: 10.1016/j.neuroscience.2023.08.033] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 08/04/2023] [Accepted: 08/27/2023] [Indexed: 09/02/2023]
Abstract
Diabetes Mellitus (DM) and Alzheimer's disease (AD) have been two of the most common chronic diseases affecting people worldwide. Type 2 DM (T2DM) is a metabolic disease depicted by insulin resistance, dyslipidemia, and chronic hyperglycemia while AD is a neurodegenerative disease marked by Amyloid β (Aβ) accumulation, neurofibrillary tangles aggregation, and tau phosphorylation. Various clinical, epidemiological, and lipidomics studies have linked those diseases claiming shared pathological pathways raising the assumption that diabetic patients are at an increased risk of developing AD later in their lives. Insulin resistance is the tipping point beyond where advanced glycation end (AGE) products and free radicals are produced leading to oxidative stress and lipid peroxidation. Additionally, different types of lipids are playing a crucial role in the development and the relationship between those diseases. Lipidomics, an analysis of lipid structure, formation, and interactions, evidently exhibits these lipid changes and their direct and indirect effect on Aβ synthesis, insulin resistance, oxidative stress, and neuroinflammation. In this review, we have discussed the pathophysiology of T2DM and AD, the interconnecting pathological pathways they share, and the lipidomics where different lipids such as cholesterol, phospholipids, sphingolipids, and sulfolipids contribute to the underlying features of both diseases. Understanding their role can be beneficial for diagnostic purposes or introducing new drugs to counter AD.
Collapse
Affiliation(s)
| | - Sarah Rahman
- School of Medicine, Tehran University of Medical Sciences, Iran
| |
Collapse
|
9
|
Lotfy MM, Dowidar MF, Ali HA, Ghonimi WAM, AL-Farga A, Ahmed AI. Effect of Selenium Nanoparticles and/or Bee Venom against STZ-Induced Diabetic Cardiomyopathy and Nephropathy. Metabolites 2023; 13:400. [PMID: 36984840 PMCID: PMC10057804 DOI: 10.3390/metabo13030400] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/03/2022] [Accepted: 02/22/2023] [Indexed: 03/11/2023] Open
Abstract
The main purpose of our study was to examine the role of selenium nanoparticles (SeNPs) and/or bee venom (BV) in ameliorating diabetic cardiomyopathy (DCM) and nephropathy (DN) at the biochemical, histopathological and molecular levels. Fifty male albino rats were used in this experiment, divided into five groups: control, Streptozocin (STZ) diabetic, STZ-diabetic treated with SeNPs, STZ-diabetic treated with BV, and STZ-diabetic treated with SeNPs and BV. Biochemically, STZ injection resulted in a significant increase in serum glucose, BUN, creatinine, CRP, CK-MB, AST, LDH and cardiac troponins with a significant decrease in the serum insulin and albumin concentrations. Histopathologically, STZ injection resulted in diabetes, as revealed by glomerulonephritis, perivascular hemorrhage, inflammatory cell infiltrations and fibrosis, with widening of interstitial spaces of cardiomyocytes, loss of muscle cells continuity and some hyaline degeneration. At the molecular levels, the expression levels of miRNA 328, miRNA-21, TGFβ1, TGFβ1R, JAK1, STST-3, SMAD-1 and NFκβ genes were significantly up-regulated, whereas the expression levels of SMAD-7 were significantly down-regulated. It is concluded that SeNPs and/or BV administration ameliorates the deleterious effects resulting from STZ administration through improving the biochemical, histopathological and molecular effects, suggesting their protective role against the long-term diabetic complications of DCM and DN.
Collapse
Affiliation(s)
- Mona M. Lotfy
- Biochemistry Departments, Faculty of Vet. Med., Zagazig University, Zagazig 44519, Egypt
| | - Mohamed F. Dowidar
- Biochemistry Departments, Faculty of Vet. Med., Zagazig University, Zagazig 44519, Egypt
| | - Haytham A. Ali
- Biochemistry Departments, Faculty of Vet. Med., Zagazig University, Zagazig 44519, Egypt
- Department of Biochemistry, College of science, University of Jeddah, Jeddah 23218, Saudi Arabia
| | - Wael A. M. Ghonimi
- Department of Histology and Cytology, Faculty of Vet. Med., Zagazig University, Zagazig 44519, Egypt
| | - Ammar AL-Farga
- Department of Biochemistry, College of science, University of Jeddah, Jeddah 23218, Saudi Arabia
| | - Amany I. Ahmed
- Biochemistry Departments, Faculty of Vet. Med., Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
10
|
Suh J, Choi Y, Oh JS, Song K, Choi HS, Kwon A, Chae HW, Kim HS. Association between early glycemic management and diabetes complications in type 1 diabetes mellitus: A retrospective cohort study. Prim Care Diabetes 2023; 17:60-67. [PMID: 36609067 DOI: 10.1016/j.pcd.2022.12.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 12/20/2022] [Accepted: 12/26/2022] [Indexed: 01/06/2023]
Abstract
AIMS To investigate the association between early HbA1c levels near diagnosis and future glycemic management, and analyzed risk factors of complications in people with T1DM. METHODS This retrospective cohort study included 201 children and adolescents with T1DM. Patient data including sex, age at diagnosis, duration of disease, HbA1c levels, HbA1c variability during the follow-up period, and diabetes complications and comorbidities were collected. RESULTS The mean follow-up period of patients was 16.4 years. HbA1c levels in all three examined time points after diagnosis (first year, second year, and first two years) were significantly associated with recent HbA1c level, and second-year HbA1c was most closely correlated with recent HbA1c level. Elevated second-year HbA1c was a risk factor of diabetic ketoacidosis (DKA) and retinopathy, and increased variability of HbA1c was significantly related to various microvascular complications. When HbA1c is stratified into quartiles, the subjects of each quartile trend to stay within that quartile over the follow-up period. CONCLUSIONS Early HbA1c levels were closely associated with recent HbA1c levels and diabetes complications in patients with T1DMs. Strict glucose management after diagnosis and reducing variability of HbA1c may prevent future diabetes complications and comorbidities.
Collapse
Affiliation(s)
- Junghwan Suh
- Department of Pediatrics, Severance Children's Hospital, Endocrine Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Youngha Choi
- Department of Pediatrics, Severance Children's Hospital, Endocrine Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Jun Suk Oh
- Department of Pediatrics, Severance Children's Hospital, Endocrine Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Kyungchul Song
- Department of Pediatrics, Severance Children's Hospital, Endocrine Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Han Saem Choi
- Department of Pediatrics, Severance Children's Hospital, Endocrine Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Ahreum Kwon
- Department of Pediatrics, Severance Children's Hospital, Endocrine Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyun Wook Chae
- Department of Pediatrics, Severance Children's Hospital, Endocrine Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Ho-Seong Kim
- Department of Pediatrics, Severance Children's Hospital, Endocrine Research Institute, Yonsei University College of Medicine, Seoul, South Korea.
| |
Collapse
|
11
|
Tanday N, Lafferty RA, Flatt PR, Irwin N. Beneficial metabolic effects of recurrent periods of beta-cell rest and stimulation using stable neuropeptide Y1 and glucagon-like peptide-1 receptor agonists. Diabetes Obes Metab 2022; 24:2353-2363. [PMID: 35848461 PMCID: PMC9804730 DOI: 10.1111/dom.14821] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/01/2022] [Accepted: 07/12/2022] [Indexed: 01/09/2023]
Abstract
AIM To examine whether sequential administration of (d-Arg35 )-sea lamprey peptide tyrosine tyrosine (1-36) (SL-PYY) and the glucagon-like peptide-1 (GLP-1) mimetic, liraglutide, has beneficial effects in diabetes. METHODS SL-PYY is an enzymatically stable neuropeptide Y1 receptor (NPY1R) agonist known to induce pancreatic beta-cell rest and improve overall beta-cell health. We employed SL-PYY and liraglutide to induce appropriate recurrent periods of beta-cell rest and stimulation, to assess therapeutic benefits in high fat fed (HFF) mice with streptozotocin (STZ)-induced insulin deficiency, namely HFF-STZ mice. RESULTS Previous studies confirm that, at a dose of 0.25 nmol/kg, liraglutide exerts bioactivity over an 8-12 hour period in mice. Initial pharmacokinetic analysis revealed that 75 nmol/kg SL-PYY yielded a similar plasma drug time profile. When SL-PYY (75 nmol/kg) and liraglutide (0.25 nmol/kg) were administered sequentially at 08:00 AM and 08:00 PM, respectively, to HFF-STZ mice for 28 days, reductions in energy intake, body weight, circulating glucose, insulin and glucagon were noted. Similarly positive, but slightly less striking, effects were also apparent with twice-daily liraglutide-only therapy. The sequential SL-PYY and liraglutide treatment also improved insulin sensitivity and glucose-induced insulin secretory responses, which was not apparent with liraglutide treatment, although benefits on glucose tolerance were mild. Interestingly, combined therapy also elevated pancreatic insulin, decreased pancreatic glucagon and enhanced the plasma insulin/glucagon ratio compared with liraglutide alone. This was not associated with an enhancement of beneficial changes in islet cell areas, proliferation or apoptosis compared with liraglutide alone, but the numbers of centrally stained glucagon-positive islet cells were reduced by sequential combination therapy. CONCLUSION These data show that NPY1R-induced intervals of beta-cell rest, combined with GLP-1R-stimulated periods of beta-cell stimulation, should be further evaluated as an effective treatment option for obesity-driven forms of diabetes.
Collapse
Affiliation(s)
- Neil Tanday
- Biomedical Sciences Research InstituteCentre for Diabetes, Ulster UniversityColeraineNorthern Ireland
| | - Ryan A. Lafferty
- Biomedical Sciences Research InstituteCentre for Diabetes, Ulster UniversityColeraineNorthern Ireland
| | - Peter R. Flatt
- Biomedical Sciences Research InstituteCentre for Diabetes, Ulster UniversityColeraineNorthern Ireland
| | - Nigel Irwin
- Biomedical Sciences Research InstituteCentre for Diabetes, Ulster UniversityColeraineNorthern Ireland
| |
Collapse
|
12
|
Goode RA, Hum JM, Kalwat MA. Therapeutic Strategies Targeting Pancreatic Islet β-Cell Proliferation, Regeneration, and Replacement. Endocrinology 2022; 164:6836713. [PMID: 36412119 PMCID: PMC9923807 DOI: 10.1210/endocr/bqac193] [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: 09/09/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022]
Abstract
Diabetes results from insufficient insulin production by pancreatic islet β-cells or a loss of β-cells themselves. Restoration of regulated insulin production is a predominant goal of translational diabetes research. Here, we provide a brief overview of recent advances in the fields of β-cell proliferation, regeneration, and replacement. The discovery of therapeutic targets and associated small molecules has been enabled by improved understanding of β-cell development and cell cycle regulation, as well as advanced high-throughput screening methodologies. Important findings in β-cell transdifferentiation, neogenesis, and stem cell differentiation have nucleated multiple promising therapeutic strategies. In particular, clinical trials are underway using in vitro-generated β-like cells from human pluripotent stem cells. Significant challenges remain for each of these strategies, but continued support for efforts in these research areas will be critical for the generation of distinct diabetes therapies.
Collapse
Affiliation(s)
- Roy A Goode
- Division of Biomedical Sciences, College of Osteopathic Medicine, Marian University, Indianapolis, IN, USA
| | - Julia M Hum
- Division of Biomedical Sciences, College of Osteopathic Medicine, Marian University, Indianapolis, IN, USA
| | - Michael A Kalwat
- Correspondence: Michael A. Kalwat, PhD, Lilly Diabetes Center of Excellence, Indiana Biosciences Research Institute, 1210 Waterway Blvd, Suite 2000, Indianapolis, IN 46202, USA. or
| |
Collapse
|
13
|
Aslanoglou D, Bertera S, Friggeri L, Sánchez-Soto M, Lee J, Xue X, Logan RW, Lane JR, Yechoor VK, McCormick PJ, Meiler J, Free RB, Sibley DR, Bottino R, Freyberg Z. Dual pancreatic adrenergic and dopaminergic signaling as a therapeutic target of bromocriptine. iScience 2022; 25:104771. [PMID: 35982797 PMCID: PMC9379584 DOI: 10.1016/j.isci.2022.104771] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 06/10/2022] [Accepted: 07/11/2022] [Indexed: 11/23/2022] Open
Abstract
Bromocriptine is approved as a diabetes therapy, yet its therapeutic mechanisms remain unclear. Though bromocriptine's actions have been mainly attributed to the stimulation of brain dopamine D2 receptors (D2R), bromocriptine also targets the pancreas. Here, we employ bromocriptine as a tool to elucidate the roles of catecholamine signaling in regulating pancreatic hormone secretion. In β-cells, bromocriptine acts on D2R and α2A-adrenergic receptor (α2A-AR) to reduce glucose-stimulated insulin secretion (GSIS). Moreover, in α-cells, bromocriptine acts via D2R to reduce glucagon secretion. α2A-AR activation by bromocriptine recruits an ensemble of G proteins with no β-arrestin2 recruitment. In contrast, D2R recruits G proteins and β-arrestin2 upon bromocriptine stimulation, demonstrating receptor-specific signaling. Docking studies reveal distinct bromocriptine binding to α2A-AR versus D2R, providing a structural basis for bromocriptine's dual actions on β-cell α2A-AR and D2R. Together, joint dopaminergic and adrenergic receptor actions on α-cell and β-cell hormone release provide a new therapeutic mechanism to improve dysglycemia.
Collapse
Affiliation(s)
- Despoina Aslanoglou
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Suzanne Bertera
- Institute of Cellular Therapeutics, Allegheny Health Network Research Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Laura Friggeri
- Department of Chemistry, Center for Structural Biology, Vanderbilt University, Nashville, TN, USA
| | - Marta Sánchez-Soto
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Jeongkyung Lee
- Diabetes and Beta Cell Biology Center, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xiangning Xue
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ryan W. Logan
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - J. Robert Lane
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen’s Medical Centre, University of Nottingham, Nottingham, UK
- Centre of Membrane Protein and Receptors, Universities of Birmingham and Nottingham, Nottingham, UK
| | - Vijay K. Yechoor
- Diabetes and Beta Cell Biology Center, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peter J. McCormick
- Centre for Endocrinology, William Harvey Research Institute, Bart’s and the London School of Medicine and Dentistry, Queen Mary, University of London, London, UK
| | - Jens Meiler
- Department of Chemistry, Center for Structural Biology, Vanderbilt University, Nashville, TN, USA
- Institute for Drug Discovery, Leipzig University Medical School, Leipzig, Germany
| | - R. Benjamin Free
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - David R. Sibley
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Rita Bottino
- Institute of Cellular Therapeutics, Allegheny Health Network Research Institute, Allegheny Health Network, Pittsburgh, PA, USA
- Imagine Pharma, Pittsburgh, PA, USA
| | - Zachary Freyberg
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Cell Biology, University of Pittsburgh, PA, USA
| |
Collapse
|
14
|
The Use of Vasopressors During Deceased Donor Pancreas Procurement Decreases the Risk of Pancreas Transplant Graft Failure. Pancreas 2022; 51:747-751. [PMID: 36395398 DOI: 10.1097/mpa.0000000000002103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVES The objective of this study was to identify the effect of various vasopressors on pancreas graft failure and patient survival. METHODS A retrospective analysis of the United Network for Organ Sharing database was performed between 2000 and 2019. Patient and graft survival rates were analyzed up to 5 years posttransplant. RESULTS The data included 17,348 pancreas transplant recipients: 12,857 simultaneous pancreas-kidney, 1440 pancreas transplant alone, and 3051 pancreas-after-kidney transplant recipients. Use of dopamine during deceased donor procurement increased graft failure by 18% (hazard ratio [HR], 1.18; P < 0.001). Absence of vasopressor caused graft failure to rise by 8% (HR, 1.08; P = 0.09). Dopamine increased the mortality rate by 37% (HR, 1.37; P < 0.001) and the absence of vasopressor increased the mortality rate by 14% (HR, 1.14; P = 0.02). Phenylephrine and norepinephrine reduced the mortality rate by 10% (HR, 0.90; P = 0.05) and 11% (HR, 0.89; P = 0.10), respectively. CONCLUSIONS The absence of vasopressor use or the use of dopamine is associated with a higher risk of both pancreas transplant graft failure and recipient mortality. The use of phenylephrine and norepinephrine reduces the risk of mortality. This information should guide deceased donor hemodynamic support management in anticipation of pancreas procurement for future transplantation.
Collapse
|
15
|
N-3 PUFA and Pregnancy Preserve C-Peptide in Women with Type 1 Diabetes Mellitus. Pharmaceutics 2021; 13:pharmaceutics13122082. [PMID: 34959363 PMCID: PMC8703519 DOI: 10.3390/pharmaceutics13122082] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/25/2021] [Accepted: 12/02/2021] [Indexed: 12/18/2022] Open
Abstract
Type 1 diabetes (T1DM) is an autoimmune disease characterized by the gradual loss of β-cell function and insulin secretion. In pregnant women with T1DM, endogenous insulin production is absent or minimal, and exogenous insulin is required to control glycemia and prevent ketoacidosis. During pregnancy, there is a partial decrease in the activity of the immune system, and there is a suppression of autoimmune diseases. These changes in pregnant women with T1DM are reflected by Langerhans islet enlargement and improved function compared to pre-pregnancy conditions. N-3 polyunsaturated fatty acids (n-3 PUFA) have a protective effect, affect β-cell preservation, and increase endogenous insulin production. Increased endogenous insulin production results in reduced daily insulin doses, better metabolic control, and adverse effects of insulin therapy, primarily hypoglycemia. Hypoglycemia affects most pregnant women with T1DM and is several times more common than that outside of pregnancy. Strict glycemic control improves the outcome of pregnancy but increases the risk of hypoglycemia and causes maternal complications, including coma and convulsions. The suppression of the immune system during pregnancy increases the concentration of C-peptide in women with T1DM, and n-3 PUFA supplements serve as the additional support for a rise in C-peptide levels through its anti-inflammatory action.
Collapse
|
16
|
Sahin GS, Lee H, Engin F. An accomplice more than a mere victim: The impact of β-cell ER stress on type 1 diabetes pathogenesis. Mol Metab 2021; 54:101365. [PMID: 34728341 PMCID: PMC8606542 DOI: 10.1016/j.molmet.2021.101365] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/23/2021] [Accepted: 10/26/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Pancreatic β-cells are the insulin factory of an organism with a mission to regulate glucose homeostasis in the body. Due to their high secretory activity, β-cells rely on a functional and intact endoplasmic reticulum (ER). Perturbations to ER homeostasis and unmitigated stress lead to β-cell dysfunction and death. Type 1 diabetes (T1D) is a chronic inflammatory disease caused by the autoimmune-mediated destruction of β-cells. Although autoimmunity is an essential component of T1D pathogenesis, accumulating evidence suggests an important role of β-cell ER stress and aberrant unfolded protein response (UPR) in disease initiation and progression. SCOPE OF REVIEW In this article, we introduce ER stress and the UPR, review β-cell ER stress in various mouse models, evaluate its involvement in inflammation, and discuss the effects of ER stress on β-cell plasticity and demise, and islet autoimmunity in T1D. We also highlight the relationship of ER stress with other stress response pathways and provide insight into ongoing clinical studies targeting ER stress and the UPR for the prevention or treatment of T1D. MAJOR CONCLUSIONS Evidence from ex vivo studies, in vivo mouse models, and tissue samples from patients suggest that β-cell ER stress and a defective UPR contribute to T1D pathogenesis. Thus, restoration of β-cell ER homeostasis at various stages of disease presents a plausible therapeutic strategy for T1D. Identifying the specific functions and regulation of each UPR sensor in β-cells and uncovering the crosstalk between stressed β-cells and immune cells during T1D progression would provide a better understanding of the molecular mechanisms of disease process, and may reveal novel targets for development of effective therapies for T1D.
Collapse
Affiliation(s)
- Gulcan Semra Sahin
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, 53706, USA
| | - Hugo Lee
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, 53706, USA
| | - Feyza Engin
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, 53706, USA; Department of Medicine, Division of Endocrinology, Diabetes & Metabolism, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, 53705, USA; Department of Cell & Regenerative Biology, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, 53705, USA.
| |
Collapse
|
17
|
Yau B, Hocking S, Andrikopoulos S, Kebede MA. Targeting the insulin granule for modulation of insulin exocytosis. Biochem Pharmacol 2021; 194:114821. [PMID: 34748819 DOI: 10.1016/j.bcp.2021.114821] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/29/2021] [Accepted: 11/02/2021] [Indexed: 02/08/2023]
Abstract
The pancreatic β-cells control insulin secretion in the body to regulate glucose homeostasis, and β-cell stress and dysfunction is characteristic of Type 2 Diabetes. Pharmacological targeting of the β-cell to increase insulin secretion is typically utilised, however, extended use of common drugs such as sulfonylureas are known to result in secondary failure. Moreover, there is evidence they may induce β-cell failure in the long term. Within β-cells, insulin secretory granules (ISG) serve as compartments to store, process and traffic insulin for exocytosis. There is now growing evidence that ISG exist in multiple populations, distinct in their protein composition, motility, age, and capacity for secretion. In this review, we discuss the implications of a heterogenous ISG population in β-cells and highlight the need for more understanding into how unique ISG populations may be targeted in anti-diabetic therapies.
Collapse
Affiliation(s)
- Belinda Yau
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia; Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia.
| | - Samantha Hocking
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia; Central Clinical School, Faculty of Medicine and Health and Department of Endocrinology Royal Prince Alfred Hospital, NSW, Australia
| | | | - Melkam A Kebede
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia; Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
18
|
Kalwat MA, Scheuner D, Rodrigues-dos-Santos K, Eizirik DL, Cobb MH. The Pancreatic ß-cell Response to Secretory Demands and Adaption to Stress. Endocrinology 2021; 162:bqab173. [PMID: 34407177 PMCID: PMC8459449 DOI: 10.1210/endocr/bqab173] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Indexed: 02/06/2023]
Abstract
Pancreatic β cells dedicate much of their protein translation capacity to producing insulin to maintain glucose homeostasis. In response to increased secretory demand, β cells can compensate by increasing insulin production capability even in the face of protracted peripheral insulin resistance. The ability to amplify insulin secretion in response to hyperglycemia is a critical facet of β-cell function, and the exact mechanisms by which this occurs have been studied for decades. To adapt to the constant and fast-changing demands for insulin production, β cells use the unfolded protein response of the endoplasmic reticulum. Failure of these compensatory mechanisms contributes to both type 1 and 2 diabetes. Additionally, studies in which β cells are "rested" by reducing endogenous insulin demand have shown promise as a therapeutic strategy that could be applied more broadly. Here, we review recent findings in β cells pertaining to the metabolic amplifying pathway, the unfolded protein response, and potential advances in therapeutics based on β-cell rest.
Collapse
Affiliation(s)
- Michael A Kalwat
- Indiana Biosciences Research Institute, Indianapolis, IN 46202, USA
| | - Donalyn Scheuner
- Indiana Biosciences Research Institute, Indianapolis, IN 46202, USA
| | | | - Decio L Eizirik
- Indiana Biosciences Research Institute, Indianapolis, IN 46202, USA
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Melanie H Cobb
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| |
Collapse
|
19
|
Lupin γ-conglutin protects against cell death induced by oxidative stress and lipotoxicity, but transiently inhibits in vitro insulin secretion by increasing K ATP channel currents. Int J Biol Macromol 2021; 187:76-90. [PMID: 34280449 DOI: 10.1016/j.ijbiomac.2021.07.088] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 07/03/2021] [Accepted: 07/13/2021] [Indexed: 02/08/2023]
Abstract
Lupin γ-conglutin beneficially modulates glycemia, but whether it protects against oxidative and lipotoxic damage remains unknown. Here, we studied the effects of γ-conglutin on cell death provoked by hydrogen peroxide and palmitate in HepG2 hepatocytes and insulin-producing MIN6 cells, and if a modulation of mitochondrial potential and reactive oxygen species (ROS) levels was involved. We also investigated how γ-conglutin influences insulin secretion and electrical activity of β-cells. The increased apoptosis of HepG2 cells exposed to hydrogen peroxide was prevented by γ-conglutin, and the viability and ROS content in γ-conglutin-treated cells was similar to that of non-exposed cells. Additionally, γ-conglutin partially protected MIN6 cells against hydrogen peroxide-induced death. This was associated with a marked reduction in ROS. No significant changes were found in the mitochondrial potential of γ-conglutin-treated cells. Besides, we observed a partial protection against lipotoxicity only in hepatocytes. Unexpectedly, we found a transient inhibition of insulin secretion, plasma membrane hyperpolarization, and higher KATP channel currents in β-cells treated with γ-conglutin. Our data show that γ-conglutin protects against cell death induced by oxidative stress or lipotoxicity by decreasing ROS and might also indicate that γ-conglutin promotes a β-cell rest, which could be useful for preventing β-cell exhaustion in chronic hyperglycemia.
Collapse
|
20
|
Laurenti MC, Matveyenko A, Vella A. Measurement of Pulsatile Insulin Secretion: Rationale and Methodology. Metabolites 2021; 11:409. [PMID: 34206296 PMCID: PMC8305896 DOI: 10.3390/metabo11070409] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/17/2021] [Accepted: 06/18/2021] [Indexed: 12/29/2022] Open
Abstract
Pancreatic β-cells are responsible for the synthesis and exocytosis of insulin in response to an increase in circulating glucose. Insulin secretion occurs in a pulsatile manner, with oscillatory pulses superimposed on a basal secretion rate. Insulin pulses are a marker of β-cell health, and secretory parameters, such as pulse amplitude, time interval and frequency distribution, are impaired in obesity, aging and type 2 diabetes. In this review, we detail the mechanisms of insulin production and β-cell synchronization that regulate pulsatile insulin secretion, and we discuss the challenges to consider when measuring fast oscillatory secretion in vivo. These include the anatomical difficulties of measuring portal vein insulin noninvasively in humans before the hormone is extracted by the liver and quickly removed from the circulation. Peripheral concentrations of insulin or C-peptide, a peptide cosecreted with insulin, can be used to estimate their secretion profile, but mathematical deconvolution is required. Parametric and nonparametric approaches to the deconvolution problem are evaluated, alongside the assumptions and trade-offs required for their application in the quantification of unknown insulin secretory rates from known peripheral concentrations. Finally, we discuss the therapeutical implication of targeting impaired pulsatile secretion and its diagnostic value as an early indicator of β-cell stress.
Collapse
Affiliation(s)
- Marcello C. Laurenti
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic, Rochester, MN 55905, USA; (M.C.L.); (A.M.)
- Biomedical Engineering and Physiology Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA
| | - Aleksey Matveyenko
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic, Rochester, MN 55905, USA; (M.C.L.); (A.M.)
| | - Adrian Vella
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic, Rochester, MN 55905, USA; (M.C.L.); (A.M.)
| |
Collapse
|
21
|
Chan JCN, Gagliardino JJ, Ilkova H, Lavalle F, Ramachandran A, Mbanya JC, Shestakova M, Dessapt-Baradez C, Chantelot JM, Aschner P. One in Seven Insulin-Treated Patients in Developing Countries Reported Poor Persistence with Insulin Therapy: Real World Evidence from the Cross-Sectional International Diabetes Management Practices Study (IDMPS). Adv Ther 2021; 38:3281-3298. [PMID: 33978906 PMCID: PMC8189989 DOI: 10.1007/s12325-021-01736-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 03/30/2021] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Although poor adherence to insulin is widely recognised, periodic discontinuation of insulin may cause more severe hyperglycaemia than poor adherence. We assessed persistence with insulin therapy in patients with type 1 (T1D) or type 2 diabetes (T2D) in developing countries and the reasons for insulin discontinuation. METHODS The International Diabetes Management Practices Study collected real-world data from developing countries in seven waves between 2005 and 2017. In Wave 7 (2016-2017), we asked adult patients with T1D and insulin-treated T2D to report whether they had ever discontinued insulin, the estimated duration of discontinuation and underlying reasons. RESULTS Among 8303 patients recruited from 24 countries by 620 physicians, 4596 were insulin-treated (T1D: 2000; T2D: 2596). In patients with T1D, 14.0% (95% CI: 12.5-15.6) reported having self-discontinued insulin for a median duration of 1.0 month (IQR: 0.5, 3.5). The respective figures in patients with T2D were 13.7% (12.4-15.1) and 2.0 months (IQR: 1.0, 6.0). The main reasons for discontinuation were impact on social life (T1D: 41.0%; T2D: 30.5%), cost of medications and test strips (T1D: 34.4%; T2D: 24.5%), fear of hypoglycaemia (T1D: 26.7%; T2D: 28.0%) and lack of support (T1D: 26.4%; T2D: 25.9%). Other factors included age < 40 years, non-university education and short disease duration (T1D: ≤ 1 year; T2D: > 1-≤ 5 years). Patients with T1D who did not perform self-monitoring of blood glucose (SMBG) or self-adjust their insulin dosage, and patients with T1D or T2D without glucose meters were less likely to persist with insulin. Nearly 50% of patients who reported poor persistence had HbA1c > 75 mmol/mol (> 9%) and > 50% of physicians recommended diabetes education programmes to improve treatment persistence. CONCLUSION In developing countries, poor persistence with insulin is common among insulin-treated patients, supporting calls for urgent actions to ensure easy access to insulin, tools for SMBG and education.
Collapse
Affiliation(s)
- Juliana C N Chan
- Department of Medicine and Therapeutics, Hong Kong Institute of Diabetes and Obesity and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, The Prince of Wales Hospital, Shatin, Hong Kong SAR, China.
| | - Juan José Gagliardino
- CENEXA, Center of Experimental and Applied Endocrinology (La Plata National University-La Plata National Scientific and Technical Research Council), La Plata, Argentina
| | - Hasan Ilkova
- Division of Endocrinology Metabolism and Diabetes, Department of Internal Medicine, Cerrahpasa Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Fernando Lavalle
- Facultad de Medicina de la Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Ambady Ramachandran
- India Diabetes Research Foundation, Dr. A. Ramachandran's Diabetes Hospitals, Chennai, India
| | - Jean Claude Mbanya
- Biotechnology Center, Doctoral School of Life Sciences, Health and Environment, Faculty of Medicine and Biomedical Sciences, University of Yaounde I, Yaounde, Cameroon
| | - Marina Shestakova
- Endocrinology Research Center, Moscow, Russia
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | | | | | - Pablo Aschner
- Javeriana University School of Medicine, Bogotá, Colombia
- San Ignacio University Hospital, Bogotá, Colombia
| |
Collapse
|
22
|
Shapey IM, Tan ZL, Gioco R, Khambalia H, Fullwood C, Yiannoullou P, Summers A, Hanley NA, Augustine T, Rutter MK, van Dellen D. Peri-transplant glycaemic control as a predictor of pancreas transplant survival. Diabetes Obes Metab 2021; 23:49-57. [PMID: 32893472 DOI: 10.1111/dom.14181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/04/2020] [Accepted: 08/20/2020] [Indexed: 11/30/2022]
Abstract
AIMS The relationship between peri-transplant glycaemic control and outcomes following pancreas transplantation is unknown. We aimed to relate peri-transplant glycaemic control to pancreas graft survival and to develop a framework for defining early graft dysfunction. METHODS Peri-transplant glycaemic control profiles over the first 5 days postoperatively were determined by an area under the curve [AUC; average daily glucose level (mmol/L) × time (days)] and the coefficient of variation of mean daily glucose levels. Peri-transplant hyperglycaemia was defined as an AUC ≥35 mmol/day/L (daily mean blood glucose ≥7 mmol/L). Risks of graft failure associated with glycaemic control and variability and peri-transplant hyperglycaemia were determined using covariate-adjusted Cox regression. RESULTS We collected 7606 glucose readings over 5 days postoperatively from 123 pancreas transplant recipients. Glucose AUC was a significant predictor of graft failure during 3.6 years of follow-up (unadjusted HR [95% confidence interval] 1.17 [1.06-1.30], P = .002). Death censored non-technical graft failure occurred in eight (10%) recipients with peri-transplant normoglycaemia, and eight (25%) recipients with peri-transplant hyperglycaemia such that hyperglycaemia predicted a 3-fold higher risk of graft failure [HR (95% confidence interval): 3.0 (1.1-8.0); P = .028]. CONCLUSION Peri-transplant hyperglycaemia is strongly associated with graft loss and could be a valuable tool guiding individualized graft monitoring and treatment. The 5-day peri-transplant glucose AUC provides a robust and responsive framework for comparing graft function.
Collapse
Affiliation(s)
- Iestyn M Shapey
- Department of Renal and Pancreas Transplantation, Manchester University NHSFT, Manchester, UK
- Division of Diabetes, Endocrinology and Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre (MAHSC), The University of Manchester, Manchester, UK
| | - Zheng L Tan
- Division of Diabetes, Endocrinology and Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre (MAHSC), The University of Manchester, Manchester, UK
| | - Rossella Gioco
- Department of Renal and Pancreas Transplantation, Manchester University NHSFT, Manchester, UK
| | - Hussein Khambalia
- Department of Renal and Pancreas Transplantation, Manchester University NHSFT, Manchester, UK
- Division of Diabetes, Endocrinology and Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre (MAHSC), The University of Manchester, Manchester, UK
| | - Catherine Fullwood
- Division of Diabetes, Endocrinology and Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre (MAHSC), The University of Manchester, Manchester, UK
- Department of Research and Innovation (Medical Statistics), Manchester University NHSFT, Manchester, UK
| | - Petros Yiannoullou
- Department of Renal and Pancreas Transplantation, Manchester University NHSFT, Manchester, UK
- Division of Diabetes, Endocrinology and Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre (MAHSC), The University of Manchester, Manchester, UK
| | - Angela Summers
- Department of Renal and Pancreas Transplantation, Manchester University NHSFT, Manchester, UK
| | - Neil A Hanley
- Department of Renal and Pancreas Transplantation, Manchester University NHSFT, Manchester, UK
| | - Titus Augustine
- Department of Renal and Pancreas Transplantation, Manchester University NHSFT, Manchester, UK
- Division of Diabetes, Endocrinology and Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre (MAHSC), The University of Manchester, Manchester, UK
| | - Martin K Rutter
- Division of Diabetes, Endocrinology and Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre (MAHSC), The University of Manchester, Manchester, UK
- Diabetes, Endocrinology and Metabolism Centre, Manchester University NHS Foundation Trust, MAHSC, Manchester, UK
| | - David van Dellen
- Department of Renal and Pancreas Transplantation, Manchester University NHSFT, Manchester, UK
- Division of Diabetes, Endocrinology and Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre (MAHSC), The University of Manchester, Manchester, UK
| |
Collapse
|
23
|
Type 2 diabetes subgroups and potential medication strategies in relation to effects on insulin resistance and beta-cell function: A step toward personalised diabetes treatment? Mol Metab 2020; 46:101158. [PMID: 33387681 PMCID: PMC8085543 DOI: 10.1016/j.molmet.2020.101158] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/16/2020] [Accepted: 12/27/2020] [Indexed: 02/07/2023] Open
Abstract
Background Type 2 diabetes is a syndrome defined by hyperglycaemia that is the result of various degrees of pancreatic β-cell failure and reduced insulin sensitivity. Although diabetes can be caused by multiple metabolic dysfunctions, most patients are defined as having either type 1 or type 2 diabetes. Recently, Ahlqvist and colleagues proposed a new method of classifying patients with adult-onset diabetes, considering the heterogenous metabolic phenotype of the disease. This new classification system could be useful for more personalised treatment based on the underlying metabolic disruption of the disease, although to date no prospective intervention studies have generated data to support such a claim. Scope of Review In this review, we first provide a short overview of the phenotype and pathogenesis of type 2 diabetes and discuss the current and new classification systems. We then review the effects of different anti-diabetic medication classes on insulin sensitivity and β-cell function and discuss future treatment strategies based on the subgroups proposed by Ahlqvist et al. Major Conclusions The proposed novel type 2 diabetes subgroups provide an interesting concept that could lead to a better understanding of the pathophysiology of the broad group of type 2 diabetes, paving the way for personalised treatment choices based on understanding the root cause of the disease. We conclude that the novel subgroups of adult-onset diabetes would benefit from anti-diabetic medications that take into account the main pathophysiology of the disease and thereby prevent end-organ damage. However, we are only beginning to address the personalised treatment of type 2 diabetes, and studies investigating the effects of current and novel drugs in subgroups with different metabolic phenotypes are needed to develop personalised treatment of the syndrome Novel subgroups of type 2 diabetes provide a concept that could lead to a better understanding of its pathophysiology. Treatment strategies would benefit from anti-diabetic medications that influence the main pathophysiology of diabetes. Here, we review different anti-diabetic medications classes affecting insulin sensitivity and β-cell function. We suggest that future treatment strategies could benefit by taking into account subgroups provided by Ahlqvist et al.
Collapse
|
24
|
Shapey IM, Summers A, Yiannoullou P, Khambalia H, Fullwood C, Hanley NA, Casey J, Forbes S, Rosenthal M, Johnson PR, Choudhary P, Bushnell J, Shaw JAM, Augustine T, Rutter MK, van Dellen D. Donor insulin use predicts beta-cell function after islet transplantation. Diabetes Obes Metab 2020; 22:1874-1879. [PMID: 32452110 DOI: 10.1111/dom.14088] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/29/2020] [Accepted: 05/10/2020] [Indexed: 01/23/2023]
Abstract
Insulin is routinely used to manage hyperglycaemia in organ donors and during the peri-transplant period in islet transplant recipients. However, it is unknown whether donor insulin use (DIU) predicts beta-cell dysfunction after islet transplantation. We reviewed data from the UK Transplant Registry and the UK Islet Transplant Consortium; all first-time transplants during 2008-2016 were included. Linear regression models determined associations between DIU, median and coefficient of variation (CV) peri-transplant glucose levels and 3-month islet graft function. In 91 islet cell transplant recipients, DIU was associated with lower islet function assessed by BETA-2 scores (β [SE] -3.5 [1.5], P = .02), higher 3-month post-transplant HbA1c levels (5.4 [2.6] mmol/mol, P = .04) and lower fasting C-peptide levels (-107.9 [46.1] pmol/l, P = .02). Glucose at 10 512 time points was recorded during the first 5 days peri-transplant: the median (IQR) daily glucose level was 7.9 (7.0-8.9) mmol/L and glucose CV was 28% (21%-35%). Neither median glucose levels nor glucose CV predicted outcomes post-transplantation. Data on DIU predicts beta-cell dysfunction 3 months after islet transplantation and could help improve donor selection and transplant outcomes.
Collapse
Affiliation(s)
- Iestyn M Shapey
- Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
- Department of Renal and Pancreatic Transplantation, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Angela Summers
- Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
- Department of Renal and Pancreatic Transplantation, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Petros Yiannoullou
- Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
- Department of Renal and Pancreatic Transplantation, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Hussein Khambalia
- Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
- Department of Renal and Pancreatic Transplantation, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Catherine Fullwood
- Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
- Department of Research and Innovation (medical statistics), Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Neil A Hanley
- Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - John Casey
- Transplant Unit, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Shareen Forbes
- Transplant Unit, Royal Infirmary of Edinburgh, Edinburgh, UK
- Endocrinology Unit, University of Edinburgh, Edinburgh, UK
| | | | - Paul Rv Johnson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | | | - James Bushnell
- Richard Bright Renal Unit, Southmead Hospital, Bristol, UK
| | - James A M Shaw
- Institute of Cellular Medicine, Newcastle University, Newcastle, UK
| | - Titus Augustine
- Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
- Department of Renal and Pancreatic Transplantation, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Martin K Rutter
- Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
- Manchester Diabetes Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - David van Dellen
- Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
- Department of Renal and Pancreatic Transplantation, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
- Department of Research and Innovation (medical statistics), Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|
25
|
Wang H, Zhang Z, Liu Y, Yang J, Zhang J, Clark C, Rodriguez DA, Amirthalingam P, Guo Y. Pre-pregnancy body mass index in mothers, birth weight and the risk of type I diabetes in their offspring: A dose-response meta-analysis of cohort studies. J Gynecol Obstet Hum Reprod 2020; 50:101921. [PMID: 32992035 DOI: 10.1016/j.jogoh.2020.101921] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 09/18/2020] [Accepted: 09/19/2020] [Indexed: 01/11/2023]
Abstract
BACKGROUND The incidence of type I diabetes among children has increased significantly and the relationship between maternal pre-pregnancy Body Mass Index (BMI), Birth weight and risk of Type 1 diabetes in children (T1DMC) is controversial. OBJECTIVE This dose-response meta-analysis was performed to investigate the association between maternal Pre-Pregnancy Body-Mass Index, Birth Weight and the Risk of Childhood Type I Diabetes. SEARCH STRATEGY A comprehensive systematic search was conducted in MEDLINE/PubMed, SCOPUS, Cochrane, and Web of Science databases from inception to April 2019. Key search terms included "body mass index" OR "Birth weight" AND "Type 1 diabetes". SELECTION CRITERIA Peer-reviewed studies that reporting association between BMI or birth weight and type I diabetes in a retrospective or prospective study by appropriate estimates such as the hazard ratio (HR), risk ratio (RR), or odds ratio (OR) and the corresponding 95 % confidence intervals (CI). DATA COLLECTION AND ANALYSIS MOOSE guidelines were followed. Data were extracted by 2 researchers, independently. Combined hazard ratios (HRs) was evaluated by DerSimonian and Laird Random-effects model. RESULTS Two studies continuing four arms with 1,209,122 participants were eligible for pre-pregnancy BMI section meta-analysis and six studies were eligible for inclusion, providing 10,340,036 participants for birth weight section meta-analysis. Pooled results demonstrated a significant association between obesity and risk of T1DMC (HR: 1.30, 95 % CI: 1.16-1.46, I2 = 7%). The combined HR (95 % CI) showed lower risk of T1DMC in low birth weight infants (HR: 0.78, 95 % CI: 0.69-0.88, I2 = 0%) and higher risk of T1DMC in the high birth weight infants versus the normal category of birth weight (HR: 1.08, 95 % CI: 1.00-1.17, I2 = 31 %). There was a significant non-linear association between birth weight and risk of T1DMC in children (Coef =-0.00032, p = 0.001). CONCLUSIONS AND RELEVANCE This systematic review and meta-analysis identified high maternal BMI and High birth weight (HBW) increase risk of childhood T1DMC.
Collapse
Affiliation(s)
- Haiyan Wang
- Obstetrics Department, Affiliated Hospital of Chengde Medical College, Chengde City, 067100, China
| | - Zhongmin Zhang
- Clinical Laboratory, Staff Hospital of Chengde Iron & Steel Group Co., LTD., Chengde City, 067102, China
| | - Yanfang Liu
- Obstetrics Department, Affiliated Hospital of Chengde Medical College, Chengde City, 067100, China
| | - Jiaqi Yang
- Department of Preventive Medicine, Chengde Medical College, Chengde City, 067100, China
| | - Jinhuan Zhang
- Obstetrics Department, Affiliated Hospital of Chengde Medical College, Chengde City, 067100, China
| | - Cain Clark
- Centre for Sport, Exercise and Life Sciences, Coventry University, Coventry, CV1 5FB, United Kingdom
| | - David Avelar Rodriguez
- Pediatric Gastroenterology and Nutrition Unit, Instituto Nacional De Pediatria, Coyoacan, Mexico
| | - Palanisamy Amirthalingam
- Department of Pharmacy Practice, Faculty of Pharmacy, University of Tabuk, P.O. Box 741, Tabuk 71491, Saudi Arabia
| | - Yanwei Guo
- Obstetrics Department, Affiliated Hospital of Chengde Medical College, Chengde City, 067100, China.
| |
Collapse
|
26
|
Abstract
PURPOSE OF REVIEW Emerging data have suggested that β-cell dysfunction may exacerbate the development and progression of type 1 diabetes (T1D). In this review, we highlight clinical and preclinical studies suggesting a role for β-cell dysfunction during the evolution of T1D and suggest agents that may promote β-cell health in T1D. RECENT FINDINGS Metabolic abnormalities exist years before development of hyperglycemia and exhibit a reproducible pattern reflecting progressive deterioration of β-cell function and increases in β-cell stress and death. Preclinical studies indicate that T1D may be prevented by modification of pathways impacting intrinsic β-cell stress and antigen presentation. Recent findings suggest that differences in metabolic phenotypes and β-cell stress may reflect differing endotypes of T1D. Multiple pathways representing potential drug targets have been identified, but most remain to be tested in human populations with preclinical disease. SUMMARY This cumulative body of work shows clear evidence that β-cell stress, dysfunction, and death are harbingers of impending T1D and likely contribute to progression of disease and insulin deficiency. Treatment with agents targeting β-cell health could augment interventions with immunomodulatory therapies but will need to be tested in intervention studies with endpoints carefully designed to capture changes in β-cell function and health.
Collapse
Affiliation(s)
- Emily K. Sims
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
- Department of Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN
- Department of Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
| | - Raghavendra G. Mirmira
- Kovler Diabetes Center and the Department of Medicine, The University of Chicago, Chicago, IL
| | - Carmella Evans-Molina
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
- Department of Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN
- Department of Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN
- Roudebush VA Medical Center, Indianapolis, IN
| |
Collapse
|
27
|
Armiyaw L, Sarcone C, Fosam A, Muniyappa R. Increased β-Cell Responsivity Independent of Insulin Sensitivity in Healthy African American Adults. J Clin Endocrinol Metab 2020; 105:5834384. [PMID: 32382759 PMCID: PMC7266075 DOI: 10.1210/clinem/dgaa234] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 04/30/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Primary insulin hypersecretion predicts type 2 diabetes (T2DM) independent of insulin resistance. Enhanced β-cell glucose responsivity contributes to insulin hypersecretion. African Americans (AAs) are at a higher risk for T2DM than non-Hispanic Whites (NHWs). Whether AAs manifest primary insulin hypersecretion is an important topic that has not been examined systematically. OBJECTIVE To examine if nondiabetic AA adults have a higher β-cell glucose responsivity compared with NHWs. METHODS Healthy nondiabetic AA (n = 18) and NHW (n=18) subjects were prospectively recruited. Indices of β-cell function, acute C-peptide secretion (X0); basal (Φ B), first-phase (Φ 1), second-phase (Φ 2), and total β-cell responsivity to glucose (Φ TOT), were derived from modeling of insulin, C-peptide, and glucose concentrations during an intravenous glucose tolerance test. Insulin sensitivity was assessed by the hyperinsulinemic-euglycemic glucose clamp technique. RESULTS Glucose disposal rate (GDR) during clamp was similar in AAs and NHWs (GDR: [AA] 12.6 ± 3.2 vs [NHW] 12.6 ± 4.2 mg/kg fat free mass +17.7/min, P = .49). Basal insulin secretion rates were similar between the groups. AA had significantly higher X0 (4423 ± 593 vs 1807 ± 176 pmol/L, P = .007), Φ 1 [377.5 ± 59.0 vs 194.5 ± 26.6 (109) P = 0.03], and Φ TOT [76.7 ± 18.3 vs 29.6 ± 4.7 (109/min), P = 0.03], with no significant ethnic differences in Φ B and Φ 2. CONCLUSIONS Independent of insulin sensitivity, AAs showed significantly higher first-phase and total β-cell responsivity than NHWs. We propose that this difference reflects increased β-cell responsivity specifically to first-phase readily releasable insulin secretion. Future studies are warranted to identify mechanisms leading to primary β-cell hypersensitivity in AAs.
Collapse
Affiliation(s)
- Latif Armiyaw
- Clinical Endocrine Section, Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Camila Sarcone
- Clinical Endocrine Section, Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Andin Fosam
- Clinical Endocrine Section, Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Ranganath Muniyappa
- Clinical Endocrine Section, Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
- Correspondence and Reprint Requests: Ranganath Muniyappa, MD, PhD, Clinical Endocrine Section, Diabetes, Endocrinology and Obesity Branch, National Institutes of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, 10 Center Drive MSC 1613, Building 10, CRC, Rm 6-3952, Bethesda, MD 20892-1613. E-mail:
| |
Collapse
|
28
|
Food texture affects glucose tolerance by altering pancreatic β-cell function in mice consuming high-fructose corn syrup. PLoS One 2020; 15:e0233797. [PMID: 32470042 PMCID: PMC7259500 DOI: 10.1371/journal.pone.0233797] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 05/12/2020] [Indexed: 12/12/2022] Open
Abstract
The incidence of metabolic diseases, such as type 2 diabetes, has increased steadily worldwide. Diet, beverages, and food texture can all markedly influence these metabolic disorders. However, the combined effects of food texture and beverages on energy metabolism remains unclear. In the present study, we examined the effect of food texture on energy metabolism in mice administered high-fructose corn syrup (HFCS). Mice were fed a soft or hard diet along with 4.2% HFCS or tap water. Body weight and total caloric intake were not affected by food texture irrespective of HFCS consumption. However, caloric intake from HFCS (i.e., drinking volume) and diet were higher and lower, respectively, in the hard food group than in the soft food group. The hard food group’s preference for HFCS was absent in case of mice treated with the μ-opioid receptor antagonist naltrexone. Despite increased HFCS consumption, blood glucose levels were lower in the hard-diet group than in the soft-diet group. In HFCS-fed mice, insulin levels after glucose stimulation and insulin content in the pancreas were higher in the hard food group than the soft food group, whereas insulin tolerance did not differ between the groups. These food texture-induced differences in glucose tolerance were not observed in mice fed tap water. Thus, food texture appears to affect glucose tolerance by influencing pancreatic β-cell function in HFCS-fed mice. These data shed light on the combined effects of eating habits and food texture on human health.
Collapse
|
29
|
Whitticar NB, Nunemaker CS. Reducing Glucokinase Activity to Enhance Insulin Secretion: A Counterintuitive Theory to Preserve Cellular Function and Glucose Homeostasis. Front Endocrinol (Lausanne) 2020; 11:378. [PMID: 32582035 PMCID: PMC7296051 DOI: 10.3389/fendo.2020.00378] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 05/12/2020] [Indexed: 12/21/2022] Open
Abstract
Pancreatic beta-cells are the only cells in the body that can synthesize and secrete insulin. Through the process of glucose-stimulated insulin secretion, beta-cells release insulin into circulation, stimulating GLUT4-dependent glucose uptake into peripheral tissue. Insulin is normally secreted in pulses that promote signaling at the liver. Long before type 2 diabetes is diagnosed, beta-cells become oversensitive to glucose, causing impaired pulsatility and overstimulation in fasting levels of glucose. The resulting hypersecretion of insulin can cause poor insulin signaling and clearance at the liver, leading to hyperinsulinemia and insulin resistance. Continued overactivity can eventually lead to beta-cell exhaustion and failure at which point type 2 diabetes begins. To prevent or reverse the negative effects of overstimulation, beta-cell activity can be reduced. Clinical studies have revealed the potential of beta-cell rest to reverse new cases of diabetes, but treatments lack durable benefits. In this perspective, we propose an intervention that reduces overactive glucokinase activity in the beta-cell. Glucokinase is known as the glucose sensor of the beta-cell due to its high control over insulin secretion. Therefore, glycolytic overactivity may be responsible for hyperinsulinemia early in the disease and can be reduced to restore normal stimulus-secretion coupling. We have previously reported that reducing glucokinase activity in prediabetic mouse islets can restore pulsatility and enhance insulin secretion. Building on this counterintuitive finding, we review the importance of pulsatile insulin secretion and highlight how normalizing glucose sensing in the beta cell during prediabetic hyperinsulinemia may restore pulsatility and improve glucose homeostasis.
Collapse
Affiliation(s)
- Nicholas B. Whitticar
- Translational Biomedical Sciences Program, Graduate College, Ohio University, Athens, OH, United States
- Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens OH, United States
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
| | - Craig S. Nunemaker
- Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens OH, United States
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
- *Correspondence: Craig S. Nunemaker
| |
Collapse
|
30
|
Tang W, Zhang B, Wang H, Li M, Wang H, Liu F, Zhu D, Bi Y. Improved skeletal muscle energy metabolism relates to the recovery of β cell function by intensive insulin therapy in drug naïve type 2 diabetes. Diabetes Metab Res Rev 2019; 35:e3177. [PMID: 31077529 DOI: 10.1002/dmrr.3177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 04/15/2019] [Accepted: 05/07/2019] [Indexed: 12/21/2022]
Abstract
AIMS Diminished energy turnover of skeletal muscle is involved in the development of type 2 diabetes. Intensive insulin therapy has been reported to maintain glycaemic control in newly diagnosed type 2 diabetes, while the underlying mechanism remains unclear. Herein, we aimed to characterize the contribution of muscular mitochondrial oxidative phosphorylation (OxPhos) activity to insulin-induced glycaemic control. MATERIALS AND METHODS There were 21 drug naïve patients with type 2 diabetes receiving continuous subcutaneous insulin infusion for 7 days. Nine nondiabetics matched for age, body mass index, and physical activity were recruited as controls. We applied 31 P magnetic resonance spectroscopy to record in vivo muscular phosphocreatine (PCr) flux in controls and diabetics before and after insulin therapy. The mitochondrial OxPhos rate was calculated as ΔPCr / Δtime during the first 50 seconds after cessation of exercise. RESULTS In drug naïve type 2 diabetes, muscular mitochondrial OxPhos rate was restored after insulin therapy. Notably, this alteration was positively associated with the improvements of 1,5-anhydroglucitol, a serum marker for glucose control over the last 1 week, as well as homeostasis model assessment of β cell function and C-peptide/glucose ratio t0 , two indices for basal insulin secretion. Furthermore, patients with diabetes family history and more severe glucotoxicity tend to achieve greater improvement in mitochondrial function by insulin. CONCLUSIONS This study provides evidence that intensive insulin therapy facilitates muscular energy metabolism in drug naïve type 2 diabetes. It correlates to the recovery of β cell function, contributing to insulin-induced glucose control.
Collapse
Affiliation(s)
- Wenjuan Tang
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - Bing Zhang
- Department of Radiology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - Huiting Wang
- Department of Radiology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - Ming Li
- Department of Radiology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - Hongdong Wang
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - Fangcen Liu
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - Dalong Zhu
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - Yan Bi
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| |
Collapse
|
31
|
Donath MY, Meier DT, Böni-Schnetzler M. Inflammation in the Pathophysiology and Therapy of Cardiometabolic Disease. Endocr Rev 2019; 40:1080-1091. [PMID: 31127805 PMCID: PMC6624792 DOI: 10.1210/er.2019-00002] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 04/15/2019] [Indexed: 12/22/2022]
Abstract
The role of chronic inflammation in the pathogenesis of type 2 diabetes mellitus and associated complications is now well established. Therapeutic interventions counteracting metabolic inflammation improve insulin secretion and action and glucose control and may prevent long-term complications. Thus, a number of anti-inflammatory drugs approved for the treatment of other inflammatory conditions are evaluated in patients with metabolic syndrome. Most advanced are clinical studies with IL-1 antagonists showing improved β-cell function and glycemia and prevention of cardiovascular diseases and heart failure. However, alternative anti-inflammatory treatments, alone or in combinations, may turn out to be more effective, depending on genetic predispositions, duration, and manifestation of the disease. Thus, there is a great need for comprehensive and well-designed clinical studies to implement anti-inflammatory drugs in the treatment of patients with metabolic syndrome and its associated conditions.
Collapse
Affiliation(s)
- Marc Y Donath
- Clinic of Endocrinology, Diabetes and Metabolism and Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Daniel T Meier
- Clinic of Endocrinology, Diabetes and Metabolism and Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Marianne Böni-Schnetzler
- Clinic of Endocrinology, Diabetes and Metabolism and Department of Biomedicine, University of Basel, Basel, Switzerland
| |
Collapse
|
32
|
Kang T, Boland BB, Alarcon C, Grimsby JS, Rhodes CJ, Larsen MR. Proteomic Analysis of Restored Insulin Production and Trafficking in Obese Diabetic Mouse Pancreatic Islets Following Euglycemia. J Proteome Res 2019; 18:3245-3258. [PMID: 31317746 DOI: 10.1021/acs.jproteome.9b00160] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
For the treatment of patients with prediabetes or diabetes, clinical evidence has emerged that β-cell function can be restored by glucose-lowering therapeutic strategies. However, little is known about the molecular mechanisms underlying this functional adaptive behavior of the pancreatic β-cell. This study examines the dynamic changes in protein expression and phosphorylation state associated with (pro)insulin production and secretory pathway function mediated by euglycemia to induce β-cell rest in obese/diabetic db/db islet β-cells. Unbiased quantitative profiling of the protein expression and phosphorylation events that occur upon β-cell adaption during the transition from hyperglycemia to euglycemia was assessed in isolated pancreatic islets from obese diabetic db/db and wild-type (WT) mice using quantitative proteomics and phosphoproteomics together with bioinformatics analysis. Dynamic changes in the expression and phosphorylation of proteins associated with pancreatic β-cell (pro)insulin production and complementary regulated-secretory pathway regulation were observed in obese diabetic db/db islets in a hyperglycemic environment, relative to WT mouse islets in a normal euglycemic environment, that resolved when isolated db/db islets were exposed to euglycemia for 12 h in vitro. By similarly treating WT islets in parallel, the effects of tissue culture could be mostly eliminated and only those changes associated with resolution by euglycemia were assessed. Among such regulated protein phosphorylation-dependent signaling events were those associated with COPII-coated vesicle-dependent ER exit, ER-to-Golgi trafficking, clathrin-coat disassembly, and a particular association for the luminal Golgi protein kinase, FAM20C, in control of distal secretory pathway trafficking, sorting, and granule biogenesis. Protein expression and especially phosphorylation play key roles in the regulation of (pro)insulin production, correlative secretory pathway trafficking, and the restoration of β-cell secretory capacity in the adaptive functional β-cell response to metabolic demand, especially that mediated by glucose.
Collapse
Affiliation(s)
- Taewook Kang
- Protein Research Group, Department of Biochemistry and Molecular Biology , University of Southern Denmark , DK-5230 Odense M , Denmark.,The Danish Diabetes Academy , 5000 Odense , Denmark
| | - Brandon B Boland
- The Kovler Diabetes Center, Department of Medicine Section of Endocrinology, Diabetes & Metabolism , University of Chicago , Chicago , Illinois 60637 , United States.,Cardiovascular, Renal and Metabolic Disease Research , MedImmune LLC , Gaithersburg , Maryland 20878 , United States
| | - Cristina Alarcon
- The Kovler Diabetes Center, Department of Medicine Section of Endocrinology, Diabetes & Metabolism , University of Chicago , Chicago , Illinois 60637 , United States
| | - Joseph S Grimsby
- Cardiovascular, Renal and Metabolic Disease Research , MedImmune LLC , Gaithersburg , Maryland 20878 , United States
| | - Christopher J Rhodes
- The Kovler Diabetes Center, Department of Medicine Section of Endocrinology, Diabetes & Metabolism , University of Chicago , Chicago , Illinois 60637 , United States.,Cardiovascular, Renal and Metabolic Disease Research , MedImmune LLC , Gaithersburg , Maryland 20878 , United States
| | - Martin R Larsen
- Protein Research Group, Department of Biochemistry and Molecular Biology , University of Southern Denmark , DK-5230 Odense M , Denmark
| |
Collapse
|
33
|
Böni-Schnetzler M, Meier DT. Islet inflammation in type 2 diabetes. Semin Immunopathol 2019; 41:501-513. [PMID: 30989320 PMCID: PMC6592966 DOI: 10.1007/s00281-019-00745-4] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 03/29/2019] [Indexed: 12/16/2022]
Abstract
Metabolic diseases including type 2 diabetes are associated with meta-inflammation. β-Cell failure is a major component of the pathogenesis of type 2 diabetes. It is now well established that increased numbers of innate immune cells, cytokines, and chemokines have detrimental effects on islets in these chronic conditions. Recently, evidence emerged which points to initially adaptive and restorative functions of inflammatory factors and immune cells in metabolism. In the following review, we provide an overview on the features of islet inflammation in diabetes and models of prediabetes. We separately emphasize what is known on islet inflammation in humans and focus on in vivo animal models and how they are used to elucidate mechanistic aspects of islet inflammation. Further, we discuss the recently emerging physiologic signaling role of cytokines during adaptation and normal function of islet cells.
Collapse
Affiliation(s)
- Marianne Böni-Schnetzler
- Endocrinology, Diabetes and Metabolism, University Hospital of Basel, 4031, Basel, Switzerland. .,Department of Biomedicine, University Hospital and University of Basel, Hebelstrasse 20, 4031, Basel, Switzerland.
| | - Daniel T Meier
- Endocrinology, Diabetes and Metabolism, University Hospital of Basel, 4031, Basel, Switzerland.,Department of Biomedicine, University Hospital and University of Basel, Hebelstrasse 20, 4031, Basel, Switzerland
| |
Collapse
|
34
|
Tang R, Zhong T, Wu C, Zhou Z, Li X. The Remission Phase in Type 1 Diabetes: Role of Hyperglycemia Rectification in Immune Modulation. Front Endocrinol (Lausanne) 2019; 10:824. [PMID: 31849842 PMCID: PMC6901662 DOI: 10.3389/fendo.2019.00824] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 11/11/2019] [Indexed: 12/16/2022] Open
Abstract
The remission phase (or honeymoon period) is a spontaneous "temporary cure stage" in type 1 diabetes course, which provides a good human model for studying β-cell protection. The exact mechanisms are still uncertain, but one of the generally recognized mechanisms is that correction of "glucotoxicity" by exogenous insulin therapy leads to "β-cell rest" and β-cell recovery. Beyond this, the remission phase is accompanied by changes in various immune cells and immune molecules, indicating downregulation of immune response, and induction of immune tolerance. The role of hyperglycemia rectification in the regulation of immune response should be emphasized because glucose metabolism is critical to maintain the normal function of immune system. Here, recent evidence of immune modulation based on the rectification of hyperglycemia from multiple aspects such as immune cells, inflammatory cytokines, biomolecules, and cell antigenicity was reviewed. It should be noteworthy that the interaction between glucose metabolism and immune plays an important role in the pathogenesis of the remission phase. The best intervention strategy may be the combination of strict glycemic control and immune modulation to protect β-cell function as early as possible.
Collapse
Affiliation(s)
- Rong Tang
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Disease, Central South University, Changsha, China
| | - Ting Zhong
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Disease, Central South University, Changsha, China
| | - Chao Wu
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Disease, Central South University, Changsha, China
| | - Zhiguang Zhou
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Disease, Central South University, Changsha, China
- *Correspondence: Zhiguang Zhou
| | - Xia Li
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Disease, Central South University, Changsha, China
- Xia Li
| |
Collapse
|
35
|
Jahan I, Corbin KL, Bogart AM, Whitticar NB, Waters CD, Schildmeyer C, Vann NW, West HL, Law NC, Wiseman JS, Nunemaker CS. Reducing Glucokinase Activity Restores Endogenous Pulsatility and Enhances Insulin Secretion in Islets From db/db Mice. Endocrinology 2018; 159:3747-3760. [PMID: 30239634 PMCID: PMC6202857 DOI: 10.1210/en.2018-00589] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 09/12/2018] [Indexed: 12/22/2022]
Abstract
An early sign of islet failure in type 2 diabetes (T2D) is the loss of normal patterns of pulsatile insulin release. Disruptions in pulsatility are associated with a left shift in glucose sensing that can cause excessive insulin release in low glucose (relative hyperinsulinemia, a hallmark of early T2D) and β-cell exhaustion, leading to inadequate insulin release during hyperglycemia. Our hypothesis was that reducing excessive glucokinase activity in diabetic islets would improve their function. Isolated mouse islets were exposed to glucose and varying concentrations of the glucokinase inhibitor d-mannoheptulose (MH) to examine changes in intracellular calcium ([Ca2+]i) and insulin secretion. Acutely exposing islets from control CD-1 mice to MH in high glucose (20 mM) dose dependently reduced the size of [Ca2+]i oscillations detected by fura-2 acetoxymethyl. Glucokinase activation in low glucose (3 mM) had the opposite effect. We then treated islets from male and female db/db mice (age, 4 to 8 weeks) and heterozygous controls overnight with 0 to 10 mM MH to determine that 1 mM MH produced optimal oscillations. We then used 1 mM MH overnight to measure [Ca2+]i and insulin simultaneously in db/db islets. MH restored oscillations and increased insulin secretion. Insulin secretion rates correlated with MH-induced increases in amplitude of [Ca2+]i oscillations (R2 = 0.57, P < 0.01, n = 10) but not with mean [Ca2+]i levels in islets (R2 = 0.05, not significant). Our findings show that correcting glucose sensing can restore proper pulsatility to diabetic islets and improved pulsatility correlates with enhanced insulin secretion.
Collapse
Affiliation(s)
- Ishrat Jahan
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio
- Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio
| | - Kathryn L Corbin
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio
- Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio
| | - Avery M Bogart
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio
- Honors Tutorial College, Ohio University, Athens, Ohio
| | - Nicholas B Whitticar
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio
| | - Christopher D Waters
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia
| | - Cara Schildmeyer
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio
- Honors Tutorial College, Ohio University, Athens, Ohio
| | - Nicholas W Vann
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia
| | - Hannah L West
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio
- Honors Tutorial College, Ohio University, Athens, Ohio
| | - Nathan C Law
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio
| | | | - Craig S Nunemaker
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio
- Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio
| |
Collapse
|
36
|
Kalwat MA, Hwang IH, Macho J, Grzemska MG, Yang JZ, McGlynn K, MacMillan JB, Cobb MH. Chromomycin A 2 potently inhibits glucose-stimulated insulin secretion from pancreatic β cells. J Gen Physiol 2018; 150:1747-1757. [PMID: 30352794 PMCID: PMC6279362 DOI: 10.1085/jgp.201812177] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 09/10/2018] [Indexed: 12/31/2022] Open
Abstract
Drugs that target insulin secretion are useful to understand β cell function and the pathogenesis of diabetes. Kalwat et al. investigate an aureolic acid that inhibits insulin secretion and reveal that it disrupts Wnt signaling, interferes with gene expression, and suppresses Ca2+ influx in β cells. Modulators of insulin secretion could be used to treat diabetes and as tools to investigate β cell regulatory pathways in order to increase our understanding of pancreatic islet function. Toward this goal, we previously used an insulin-linked luciferase that is cosecreted with insulin in MIN6 β cells to perform a high-throughput screen of natural products for chronic effects on glucose-stimulated insulin secretion. In this study, using multiple phenotypic analyses, we found that one of the top natural product hits, chromomycin A2 (CMA2), potently inhibited insulin secretion by at least three potential mechanisms: disruption of Wnt signaling, interference of β cell gene expression, and partial suppression of Ca2+ influx. Chronic treatment with CMA2 largely ablated glucose-stimulated insulin secretion even after washout, but it did not inhibit glucose-stimulated generation of ATP or Ca2+ influx. However, by using the KATP channel opener diazoxide, we uncovered defects in depolarization-induced Ca2+ influx that may contribute to the suppressed secretory response. Glucose-responsive ERK1/2 and S6 phosphorylation were also disrupted by chronic CMA2 treatment. By querying the FUSION bioinformatic database, we revealed that the phenotypic effects of CMA2 cluster with a number of Wnt–GSK3 pathway-related genes. Furthermore, CMA2 consistently decreased GSK3β phosphorylation and suppressed activation of a β-catenin activity reporter. CMA2 and a related compound, mithramycin, are known to have DNA interaction properties, possibly abrogating transcription factor binding to critical β cell gene promoters. We observed that CMA2 but not mithramycin suppressed expression of PDX1 and UCN3. However, neither expression of INSI/II nor insulin content was affected by chronic CMA2. The mechanisms of CMA2-induced insulin secretion defects may involve components both proximal and distal to Ca2+ influx. Therefore, CMA2 is an example of a chemical that can simultaneously disrupt β cell function through both noncytotoxic and cytotoxic mechanisms. Future therapeutic applications of CMA2 and similar aureolic acid analogues should consider their potential effects on pancreatic islet function.
Collapse
Affiliation(s)
- Michael A Kalwat
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX
| | - In Hyun Hwang
- Department of Pharmacy, Woosuk University, Wanju, South Korea
| | - Jocelyn Macho
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, Santa Cruz, CA
| | - Magdalena G Grzemska
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Jonathan Z Yang
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Kathleen McGlynn
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX
| | - John B MacMillan
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, Santa Cruz, CA
| | - Melanie H Cobb
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
37
|
Guess ND. Dietary Interventions for the Prevention of Type 2 Diabetes in High-Risk Groups: Current State of Evidence and Future Research Needs. Nutrients 2018; 10:E1245. [PMID: 30200572 PMCID: PMC6163866 DOI: 10.3390/nu10091245] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 08/28/2018] [Accepted: 08/30/2018] [Indexed: 12/13/2022] Open
Abstract
A series of large-scale randomised controlled trials have demonstrated the effectiveness of lifestyle change in preventing type 2 diabetes in people with impaired glucose tolerance. Participants in these trials consumed a low-fat diet, lost a moderate amount of weight and/or increased their physical activity. Weight loss appears to be the primary driver of type 2 diabetes risk reduction, with individual dietary components playing a minor role. The effect of weight loss via other dietary approaches, such as low-carbohydrate diets, a Mediterranean dietary pattern, intermittent fasting or very-low-energy diets, on the incidence of type 2 diabetes has not been tested. These diets-as described here-could be equally, if not more effective in preventing type 2 diabetes than the tested low-fat diet, and if so, would increase choice for patients. There is also a need to understand the effect of foods and diets on beta-cell function, as the available evidence suggests moderate weight loss, as achieved in the diabetes prevention trials, improves insulin sensitivity but not beta-cell function. Finally, prediabetes is an umbrella term for different prediabetic states, each with distinct underlying pathophysiology. The limited data available question whether moderate weight loss is effective at preventing type 2 diabetes in each of the prediabetes subtypes.
Collapse
Affiliation(s)
- Nicola D Guess
- Department of Nutritional Sciences, King's College London, 150 Stamford Street, Room 4.13, London SE1 9NH, UK.
| |
Collapse
|
38
|
Hasib A, Ng MT, Khan D, Gault VA, Flatt PR, Irwin N. Characterisation and antidiabetic utility of a novel hybrid peptide, exendin-4/gastrin/xenin-8-Gln. Eur J Pharmacol 2018; 834:126-135. [DOI: 10.1016/j.ejphar.2018.07.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 06/29/2018] [Accepted: 07/13/2018] [Indexed: 12/14/2022]
|
39
|
Fitas AL, Martins C, Borrego LM, Lopes L, Jörns A, Lenzen S, Limbert C. Immune cell and cytokine patterns in children with type 1 diabetes mellitus undergoing a remission phase: A longitudinal study. Pediatr Diabetes 2018. [PMID: 29527790 DOI: 10.1111/pedi.12671] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE Type 1 diabetes (T1D) develops in distinct stages, before and after disease onset. Whether the natural course translates into different immunologic patterns is still uncertain. This study aimed at identifying peripheral immune patterns at key time-points, in T1D children undergoing remission phase. METHODS Children with new-onset T1D and healthy age and gender-matched controls were recruited at a pediatric hospital. Peripheral blood samples were evaluated by flow cytometry at 3 longitudinal time-points: onset (T1), remission phase (T2) and established disease (T3). Cytokine levels were quantified by multiplex assay. Fasting C-peptide, HbA1c, and 25OHD were also measured. RESULTS T1D children (n = 28; 10.0 ± 2.6 years) showed significant differences from controls in circulating neutrophils, T helper (Th)17 and natural killer (NK) cells, with relevant variations during disease progression. At onset, neutrophils, NK, Th17 and T cytotoxic (Tc)17 cells were decreased. As disease progressed, neutrophil counts recovered whereas NK counts remained low. Th17 and Tc17 cells behavior followed the neutrophil variation pattern. B-cells were lowest in the remission phase and regulatory T-cells significantly declined after remission. Two cytokine response profiles were identified. Low cytokine-responders showed higher circulating fasting C-peptide levels at onset and longer remission periods. C-peptide inversely correlated with pro-inflammatory and cytotoxic cells. CONCLUSIONS Our data suggest an association between immune cells, cytokine patterns and metabolic counterparts. The dynamic changes of circulating immune cells during disease progression involve key innate and acquired immune cell types. This longitudinal picture of T1D progression may enable disease staging and patient stratification, essential for individualized treatment.
Collapse
Affiliation(s)
- Ana Laura Fitas
- Paediatric Endocrinology Unit, Hospital de Dona Estefânia, Centro Hospitalar de Lisboa Central, Lisbon, Portugal
| | - Catarina Martins
- Chronic Diseases Research Center CEDOC-NOVA Medical School, Lisbon, Portugal
| | - Luís Miguel Borrego
- Chronic Diseases Research Center CEDOC-NOVA Medical School, Lisbon, Portugal
| | - Lurdes Lopes
- Paediatric Endocrinology Unit, Hospital de Dona Estefânia, Centro Hospitalar de Lisboa Central, Lisbon, Portugal
| | - Anne Jörns
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Sigurd Lenzen
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany.,Institute of Experimental Diabetes Research, Hannover Medical School, Hannover, Germany
| | - Catarina Limbert
- Paediatric Endocrinology Unit, Hospital de Dona Estefânia, Centro Hospitalar de Lisboa Central, Lisbon, Portugal
| |
Collapse
|
40
|
Johnson SR, McGown I, Oppermann U, Conwell LS, Harris M, Duncan EL. A novel INS mutation in a family with maturity-onset diabetes of the young: Variable insulin secretion and putative mechanisms. Pediatr Diabetes 2018; 19:905-909. [PMID: 29633446 DOI: 10.1111/pedi.12679] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 03/01/2018] [Accepted: 03/22/2018] [Indexed: 02/06/2023] Open
Abstract
Insulin gene (INS) mutations cause a rare form of maturity-onset diabetes of the young (MODY), a heterogeneous group of autosomal dominant diabetes with at least 14 confirmed causative genes. Here, we describe a family with MODY due to a novel INS mutation, detected using massively parallel sequencing (MPS). The proband presented aged 11 years with mild diabetic ketoacidosis. She was negative for IA2 and GAD antibodies. She had a strong family history of diabetes affecting both her two siblings and her mother, none of whom had ketosis but who were considered to have type 1 diabetes and managed on insulin, and her maternal grandfather, who was managed for decades on sulfonylureas. Of note, her younger sister had insulin deficiency but an elevated fasting proinsulin:insulin ratio of 76% (ref 5%-30%). Sanger sequencing of HNF4A, HNF1A, and HNF1B in the proband was negative. Targeted MPS using a custom-designed amplicon panel sequenced on an Illumina MiSeq detected a heterozygous INS mutation c.277G>A (p.Glu93Lys). Sanger sequencing confirmed the variant segregated with diabetes within the family. Structural analysis of this variant suggested disruption of a critical hydrogen bond between insulin and the insulin receptor; however, the clinical picture in some individuals also suggested abnormal insulin processing and insulin deficiency. This family has a novel INS mutation and demonstrated variable insulin deficiency. MPS represents an efficient method of MODY diagnosis in families with rarer gene mutations.
Collapse
Affiliation(s)
- Stephanie R Johnson
- Department of Endocrinology, Lady Cilento Children's Hospital, South Brisbane, QLD, Australia.,Faculty of Medicine, The University of Queensland, Herston, QLD, Australia.,Institute of Health and Biomedical Innovation, Faculty of Health, Queensland University of Technology, Translational Research Institute, Princess Alexandra Hospital, Woolloongabba, QLD, Australia
| | - Ivan McGown
- Molecular Genetics, Mater Health Services, South Brisbane, QLD, Australia
| | - Udo Oppermann
- Structural Genomics Consortium, Botnar Research Center, Oxford Biomedical Research Unit, University of Oxford, Oxford, UK.,Freiburg Institute of Advanced Studies (FRIAS), University of Freiburg, Freiburg, Germany
| | - Louise S Conwell
- Department of Endocrinology, Lady Cilento Children's Hospital, South Brisbane, QLD, Australia.,Faculty of Medicine, The University of Queensland, Herston, QLD, Australia
| | - Mark Harris
- Department of Endocrinology, Lady Cilento Children's Hospital, South Brisbane, QLD, Australia.,Faculty of Medicine, The University of Queensland, Herston, QLD, Australia.,University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Emma L Duncan
- Faculty of Medicine, The University of Queensland, Herston, QLD, Australia.,Institute of Health and Biomedical Innovation, Faculty of Health, Queensland University of Technology, Translational Research Institute, Princess Alexandra Hospital, Woolloongabba, QLD, Australia.,Department of Endocrinology, Royal Brisbane & Women's Hospital, Herston, QLD, Australia
| |
Collapse
|
41
|
Chon S, Rhee SY, Ahn KJ, Baik SH, Park Y, Nam MS, Lee KW, Yoo SJ, Koh G, Lee DH, Kim YS, Woo JT. Long-term effects on glycaemic control and β-cell preservation of early intensive treatment in patients with newly diagnosed type 2 diabetes: A multicentre randomized trial. Diabetes Obes Metab 2018; 20:1121-1130. [PMID: 29272062 DOI: 10.1111/dom.13196] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 12/11/2017] [Accepted: 12/18/2017] [Indexed: 12/16/2022]
Abstract
AIM To determine the effects of early intensive glycaemic control with intensive insulin treatment (IIT) or initial combined oral antidiabetic drug (COAD) therapy on long-term glycaemic control and the preservation of β-cell function in people with type 2 diabetes mellitus (T2DM). METHODS Newly diagnosed drug-naïve patients with T2DM from 8 outpatient diabetes centres were randomized to receive either IIT (n = 50; glargine/glulisine) or COAD (n = 47; glimepiride/metformin) as intensive treatment until the termination criteria to ensure euglycaemia were met. After intensive treatment, the patients completed a follow-up period with either lifestyle modification (LSM) alone or rescue therapy to maintain target glycated haemoglobin levels of <7% (53 mmol/mol) up to week 104. The primary outcomes were analysed after excluding participants who were anti-glutamic acid decarboxylase autoantibody-positive. RESULTS Both intensive treatment methods were effective for short-term glycaemic control, but improvements in the disposition index (DI) were significantly greater in the IIT group than in the COAD group (P = .021). During the follow-up period after intensive treatment, the two groups significantly differed in rescue method regarding the maintenance of comparable levels of glycaemic control (P = .010) and more participants who received IIT exhibited well-controlled glycaemia with LSM alone. Additionally, the IIT group maintained a higher DI than the COAD group during the follow-up period. Cox regression analysis showed that the IIT method was associated with a 52.5% lower risk of failing to maintain drug-free glycaemic remission compared with the COAD method (P = .015). CONCLUSIONS The findings indicate that outpatient clinic-based IIT to ensure euglycaemia in newly diagnosed patients with T2DM might be an effective initial therapeutic option for improvements in β-cell function and glycaemic control over the long term, without serious adverse events.
Collapse
Affiliation(s)
- Suk Chon
- Department of Endocrinology and Metabolism, Kyung Hee University School of Medicine, Seoul, Korea
| | - Sang Youl Rhee
- Department of Endocrinology and Metabolism, Kyung Hee University School of Medicine, Seoul, Korea
| | - Kyu Jeung Ahn
- Department of Endocrinology and Metabolism, Kyung Hee University School of Medicine, Seoul, Korea
| | - Sei Hyun Baik
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Yongsoo Park
- Department of Internal Medicine, Hanyang University College of Medicine, Guri, Korea
| | - Moon Suk Nam
- Department of Internal Medicine, Inha University School of Medicine, Incheon, Korea
| | - Kwan Woo Lee
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Korea
| | - Soon Jib Yoo
- Department of Endocrinology and Metabolism, Bucheon St. Mary's Hospital, The Catholic University of Korea, Bucheon, Korea
| | - Gwanpyo Koh
- Department of Internal Medicine, Jeju National University School of Medicine, Jeju, Korea
| | - Dae Ho Lee
- Department of Internal Medicine, Gachon University Gil Medical Center, Incheon, Korea
| | - Young Seol Kim
- Department of Endocrinology and Metabolism, Kyung Hee University School of Medicine, Seoul, Korea
| | - Jeong-Taek Woo
- Department of Endocrinology and Metabolism, Kyung Hee University School of Medicine, Seoul, Korea
| |
Collapse
|
42
|
Pancreatic Islet Blood Flow Dynamics in Primates. Cell Rep 2018; 20:1490-1501. [PMID: 28793270 PMCID: PMC5575201 DOI: 10.1016/j.celrep.2017.07.039] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 05/05/2017] [Accepted: 07/14/2017] [Indexed: 12/26/2022] Open
Abstract
Blood flow regulation in pancreatic islets is critical for function but poorly understood. Here, we establish an in vivo imaging platform in a non-human primate where islets transplanted autologously into the anterior chamber of the eye are monitored non-invasively and longitudinally at single-cell resolution. Engrafted islets were vascularized and innervated and maintained the cytoarchitecture of in situ islets in the pancreas. Blood flow velocity in the engrafted islets was not affected by increasing blood glucose levels and/or the GLP-1R agonist liraglutide. However, islet blood flow was dynamic in nature and fluctuated in various capillaries. This was associated with vasoconstriction events resembling a sphincter-like action, most likely regulated by adrenergic signaling. These observations suggest a mechanism in primate islets that diverts blood flow to cell regions with higher metabolic demand. The described imaging technology applied in non-human primate islets may contribute to a better understanding of human islet pathophysiology. Monkey islets transplanted autologously into the anterior chamber of the eye (ACE) Monkey ACE islets imaged in vivo, longitudinally, and at single-cell resolution Monkey islet blood flow is dynamic and unaffected by glucose/liraglutide treatment Directional blood flow may be explained by islet structure-function relationship
Collapse
|
43
|
Neelankal John A, Jiang FX. An overview of type 2 diabetes and importance of vitamin D3-vitamin D receptor interaction in pancreatic β-cells. J Diabetes Complications 2018; 32:429-443. [PMID: 29422234 DOI: 10.1016/j.jdiacomp.2017.12.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 12/03/2017] [Accepted: 12/07/2017] [Indexed: 02/07/2023]
Abstract
One significant health issue that plagues contemporary society is that of Type 2 diabetes (T2D). This disease is characterised by higher-than-average blood glucose levels as a result of a combination of insulin resistance and insufficient insulin secretions from the β-cells of pancreatic islets of Langerhans. Previous developmental research into the pancreas has identified how early precursor genes of pancreatic β-cells, such as Cpal, Ngn3, NeuroD, Ptf1a, and cMyc, play an essential role in the differentiation of these cells. Furthermore, β-cell molecular characterization has also revealed the specific role of β-cell-markers, such as Glut2, MafA, Ins1, Ins2, and Pdx1 in insulin expression. The expression of these genes appears to be suppressed in the T2D β-cells, along with the reappearance of the early endocrine marker genes. Glucose transporters transport glucose into β-cells, thereby controlling insulin release during hyperglycaemia. This stimulates glycolysis through rises in intracellular calcium (a process enhanced by vitamin D) (Norman et al., 1980), activating 2 of 4 proteinases. The rise in calcium activates half of pancreatic β-cell proinsulinases, thus releasing free insulin from granules. The synthesis of ATP from glucose by glycolysis, Krebs cycle and oxidative phosphorylation plays a role in insulin release. Some studies have found that the β-cells contain high levels of the vitamin D receptor; however, the role that this plays in maintaining the maturity of the β-cells remains unknown. Further research is required to develop a more in-depth understanding of the role VDR plays in β-cell function and the processes by which the beta cell function is preserved.
Collapse
Affiliation(s)
- Abraham Neelankal John
- Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands, Western Australia, Australia; School of Medicine and Pharmacology, University of Western Australia, Carwley, Western Australia, Australia
| | - Fang-Xu Jiang
- Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands, Western Australia, Australia; School of Medicine and Pharmacology, University of Western Australia, Carwley, Western Australia, Australia.
| |
Collapse
|
44
|
Khan D, Moffet CR, Flatt PR, Kelly C. Role of islet peptides in beta cell regulation and type 2 diabetes therapy. Peptides 2018; 100:212-218. [PMID: 29412821 DOI: 10.1016/j.peptides.2017.11.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 11/22/2017] [Accepted: 11/22/2017] [Indexed: 12/25/2022]
Abstract
The endocrine pancreas is composed of islets of Langerhans, which secrete a variety of peptide hormones critical for the maintenance of glucose homeostasis. Insulin is the primary regulator of glucose and its secretion from beta-cells is tightly regulated in response to physiological demands. Direct cell-cell communication within islets is essential for glucose-induced insulin secretion. Emerging data suggest that islet connectivity is also important in the regulating the release of other islet hormones including glucagon and somatostatin. Autocrine and paracrine signals exerted by secreted peptides within the islet also play a key role. A great deal of attention has focused on classical islet peptides, namely insulin, glucagon and somatostatin. Recently, it has become clear that islets also synthesise and secrete a range of non-classical peptides, which regulate beta-cell function and insulin release. The current review summarises the roles of islet cell connectivity and islet peptide-driven autocrine and paracrine signalling in beta-cell function and survival. The potential to harness the paracrine effects of non-classical islet peptides for the treatment of type 2 diabetes is also briefly discussed.
Collapse
Affiliation(s)
- Dawood Khan
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute, University of Ulster, C-TRIC Building, Altnagelvin Area Hospital, Glenshane Road, L/Derry, BT47 6SB, Northern Ireland, UK
| | - Charlotte R Moffet
- SAAD Centre for Pharmacy and Diabetes, School of Biomedical Sciences, University of Ulster, Coleraine, Northern Ireland, UK
| | - Peter R Flatt
- SAAD Centre for Pharmacy and Diabetes, School of Biomedical Sciences, University of Ulster, Coleraine, Northern Ireland, UK
| | - Catriona Kelly
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute, University of Ulster, C-TRIC Building, Altnagelvin Area Hospital, Glenshane Road, L/Derry, BT47 6SB, Northern Ireland, UK.
| |
Collapse
|
45
|
Hasib A, Ng MT, Khan D, Gault VA, Flatt PR, Irwin N. A novel GLP-1/xenin hybrid peptide improves glucose homeostasis, circulating lipids and restores GIP sensitivity in high fat fed mice. Peptides 2018; 100:202-211. [PMID: 29412820 DOI: 10.1016/j.peptides.2017.10.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 10/24/2017] [Accepted: 10/24/2017] [Indexed: 02/08/2023]
Abstract
Combined modulation of peptide hormone receptors including, glucagon-like peptide-1 (GLP-1), glucose-dependent insulinotropic polypeptide (GIP) and xenin, have established benefits for the treatment of diabetes. The present study has assessed the biological actions and therapeutic efficacy of a novel exendin-4/xenin-8-Gln hybrid peptide, both alone and in combination with the GIP receptor agonist (DAla2)GIP. Exendin-4/xenin-8-Gln was enzymatically stable and exhibited enhanced insulin secretory actions when compared to its parent peptides. Exendin-4/xenin-8-Gln also possessed ability to potentiate the in vitro actions of GIP. Acute administration of exendin-4/xenin-8-Gln in mice induced appetite suppressive effects, as well as significant and protracted glucose-lowering and insulin secretory actions. Twice daily administration of exendin-4/xenin-8-Gln, alone or in combination with (DAla2)GIP, for 21-days significantly reduced non-fasting glucose and increased circulating insulin levels in high fat fed mice. In addition, all exendin-4/xenin-8-Gln treated mice displayed improved glucose tolerance, insulin sensitivity and metabolic responses to GIP. Combination therapy with (DAla2)GIP did not result in any obvious further benefits. Metabolic improvements in all treatment groups were accompanied by reduced pancreatic beta-cell area and insulin content, suggesting reduced insulin demand. Interestingly, body weight, food intake, circulating glucagon, metabolic rate and amylase activity were unaltered by the treatment regimens. However, all treatment groups, barring (DAla2)GIP alone, exhibited marked reductions in total- and LDL-cholesterol. Furthermore, exendin-4 therapy also reduced circulating triacylglycerol. This study highlights the positive antidiabetic effects of exendin-4/xenin-8-Gln, and suggests that combined modulation of GLP-1 and xenin related signalling pathways represents an exciting treatment option for type 2 diabetes.
Collapse
Affiliation(s)
- Annie Hasib
- From the SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, Northern Ireland, UK
| | - Ming T Ng
- From the SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, Northern Ireland, UK
| | - Dawood Khan
- From the SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, Northern Ireland, UK
| | - Victor A Gault
- From the SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, Northern Ireland, UK
| | - Peter R Flatt
- From the SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, Northern Ireland, UK
| | - Nigel Irwin
- From the SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, Northern Ireland, UK.
| |
Collapse
|
46
|
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: 55] [Impact Index Per Article: 7.9] [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
|
47
|
Sabatini PV, Speckmann T, Nian C, Glavas MM, Wong CK, Yoon JS, Kin T, Shapiro AMJ, Gibson WT, Verchere CB, Lynn FC. Neuronal PAS Domain Protein 4 Suppression of Oxygen Sensing Optimizes Metabolism during Excitation of Neuroendocrine Cells. Cell Rep 2018; 22:163-174. [PMID: 29298418 DOI: 10.1016/j.celrep.2017.12.033] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 10/27/2017] [Accepted: 12/08/2017] [Indexed: 02/07/2023] Open
Abstract
Depolarization of neuroendocrine cells results in calcium influx, which induces vesicle exocytosis and alters gene expression. These processes, along with the restoration of resting membrane potential, are energy intensive. We hypothesized that cellular mechanisms exist to maximize energy production during excitation. Here, we demonstrate that NPAS4, an immediate early basic helix-loop-helix (bHLH)-PAS transcription factor, acts to maximize energy production by suppressing hypoxia-inducible factor 1α (HIF1α). As such, knockout of Npas4 from insulin-producing β cells results in reduced OXPHOS, loss of insulin secretion, β cell dedifferentiation, and type 2 diabetes. NPAS4 plays a similar role in the nutrient-sensing cells of the hypothalamus. Its knockout here results in increased food intake, reduced locomotor activity, and elevated peripheral glucose production. In conclusion, NPAS4 is critical for the coordination of metabolism during the stimulation of electrically excitable cells; its loss leads to the defects in cellular metabolism that underlie the cellular dysfunction that occurs in metabolic disease.
Collapse
Affiliation(s)
- Paul V Sabatini
- Diabetes Research Group, BC Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Thilo Speckmann
- Diabetes Research Group, BC Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Cuilan Nian
- Diabetes Research Group, BC Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Maria M Glavas
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Chi Kin Wong
- Diabetes Research Group, BC Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Ji Soo Yoon
- Diabetes Research Group, BC Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Tatsuya Kin
- Department of Surgery and Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - A M James Shapiro
- Department of Surgery and Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - William T Gibson
- Diabetes Research Group, BC Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - C Bruce Verchere
- Diabetes Research Group, BC Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Francis C Lynn
- Diabetes Research Group, BC Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Surgery, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
48
|
Valitsky M, Hoffman A, Unterman T, Bar-Tana J. Insulin sensitizer prevents and ameliorates experimental type 1 diabetes. Am J Physiol Endocrinol Metab 2017; 313:E672-E680. [PMID: 28270441 DOI: 10.1152/ajpendo.00329.2016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 02/27/2017] [Accepted: 03/01/2017] [Indexed: 01/12/2023]
Abstract
Insulin-dependent type-1 diabetes (T1D) is driven by autoimmune β-cell failure, whereas systemic resistance to insulin is considered the hallmark of insulin-independent type-2 diabetes (T2D). In contrast to this canonical dichotomy, insulin resistance appears to precede the overt diabetic stage of T1D and predict its progression, implying that insulin sensitizers may change the course of T1D. However, previous attempts to ameliorate T1D in animal models or patients by insulin sensitizers have largely failed. Sensitization to insulin by MEthyl-substituted long-chain DICArboxylic acid (MEDICA) analogs in T2D animal models surpasses that of current insulin sensitizers, thus prompting our interest in probing MEDICA in the T1D context. MEDICA efficacy in modulating the course of T1D was verified in streptozotocin (STZ) diabetic rats and autoimmune nonobese diabetic (NOD) mice. MEDICA treatment normalizes overt diabetes in STZ diabetic rats when added on to subtherapeutic insulin, and prevents/delays autoimmune T1D in NOD mice. MEDICA treatment does not improve β-cell insulin content or insulitis score, but its efficacy is accounted for by pronounced total body sensitization to insulin. In conclusion, potent insulin sensitizers may counteract genetic predisposition to autoimmune T1D and amplify subtherapeutic insulin into an effective therapeutic measure for the treatment of overt T1D.
Collapse
Affiliation(s)
- Michael Valitsky
- Department of Human Nutrition and Metabolism, Hebrew University Medical School, Jerusalem, Israel
| | - Amnon Hoffman
- Institute for Drug Research, Hebrew University Faculty of Medicine, Jerusalem, Israel; and
| | - Terry Unterman
- Section of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago College of Medicine, Chicago, Illinois
| | - Jacob Bar-Tana
- Department of Human Nutrition and Metabolism, Hebrew University Medical School, Jerusalem, Israel;
| |
Collapse
|
49
|
Chemistry and biology of reactive species with special reference to the antioxidative defence status in pancreatic β-cells. Biochim Biophys Acta Gen Subj 2017; 1861:1929-1942. [PMID: 28527893 DOI: 10.1016/j.bbagen.2017.05.013] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 05/12/2017] [Accepted: 05/16/2017] [Indexed: 01/08/2023]
Abstract
BACKGROUND Diabetes mellitus is a serious metabolic disease. Dysfunction and subsequent loss of the β-cells in the islets of Langerhans through apoptosis ultimately cause a life-threatening insulin deficiency. The underlying reason for the particular vulnerability of the β-cells is an extraordinary sensitivity to the toxicity of reactive oxygen and nitrogen species (ROS and RNS) due to its low antioxidative defense status. SCOPE REVIEW This review considers the different aspects of the chemistry and biology of the biologically most important reactive species and their chemico-biological interactions in the β-cell toxicity of proinflammatory cytokines in type 1 diabetes and of lipotoxicity in type 2 diabetes development. MAJOR CONCLUSION The weak antioxidative defense equipment in the different subcellular organelles makes the β-cells particularly vulnerable and prone to mitochondrial, peroxisomal and ER stress. Looking upon the enzyme deficiencies which are responsible for the low antioxidative defense status of the pancreatic β-cells it is the lack of enzymatic capacity for H2O2 inactivation at all major subcellular sites. GENERAL SIGNIFICANCE Diabetes is the most prevalent metabolic disorder with a steadily increasing incidence of both type 1 and type 2 diabetes worldwide. The weak protection of the pancreatic β-cells against oxidative stress is a major reason for their particular vulnerability. Thus, careful protection of the β-cells is required for prevention of the disease.
Collapse
|
50
|
Khan D, Vasu S, Moffett RC, Irwin N, Flatt PR. Islet distribution of Peptide YY and its regulatory role in primary mouse islets and immortalised rodent and human beta-cell function and survival. Mol Cell Endocrinol 2016; 436:102-13. [PMID: 27465830 DOI: 10.1016/j.mce.2016.07.020] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 07/18/2016] [Accepted: 07/19/2016] [Indexed: 12/25/2022]
Abstract
Recent evidence suggests that the classic gut peptide, Peptide YY (PYY), could play a fundamental role in endocrine pancreatic function. In the present study expression of PYY and its NPY receptors on mouse islets and immortalised rodent and human beta-cells was examined together with the effects of both major circulating forms of PYY, namely PYY(1-36) and PYY(3-36), on beta-cell function, murine islet adaptions to insulin deficiency/resistance, as well as direct effects on cultured beta-cell proliferation and apoptosis. In vivo administration of PYY(3-36), but not PYY(1-36), markedly (p < 0.05) decreased food intake in overnight fasted mice. Neither form of PYY affected glucose disposal or insulin secretion following an i.p. glucose challenge. However, in vitro, PYY(1-36) and PYY(3-36) inhibited (p < 0.05 to p < 0.001) glucose, alanine and GLP-1 stimulated insulin secretion from immortalised rodent and human beta-cells, as well as isolated mouse islets, by impeding alterations in membrane potential, [Ca(2+)]i and elevations of cAMP. Mice treated with multiple low dose streptozotocin presented with severe (p < 0.01) loss of beta-cell mass accompanied by notable increases (p < 0.001) in alpha and PP cell numbers. In contrast, hydrocortisone-induced insulin resistance increased islet number (p < 0.01) and beta-cell mass (p < 0.001). PYY expression was consistently observed in alpha-, PP- and delta-, but not beta-cells. Streptozotocin decreased islet PYY co-localisation with PP (p < 0.05) and somatostatin (p < 0.001), whilst hydrocortisone increased PYY co-localisation with glucagon (p < 0.05) in mice. More detailed in vitro investigations revealed that both forms of PYY augmented (p < 0.05 to p < 0.01) immortalised human and rodent beta-cell proliferation and protected against streptozotocin-induced cytotoxicity, to a similar or superior extent as the well characterised beta-cell proliferative and anti-apoptotic agent GLP-1. Taken together, these data highlight the significance and potential offered by modulation of pancreatic islet NPY receptor signalling pathways for preservation of beta-cell mass in diabetes.
Collapse
Affiliation(s)
- Dawood Khan
- SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, Northern Ireland, UK
| | - Srividya Vasu
- SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, Northern Ireland, UK
| | - R Charlotte Moffett
- SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, Northern Ireland, UK
| | - Nigel Irwin
- SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, Northern Ireland, UK.
| | - Peter R Flatt
- SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, Northern Ireland, UK
| |
Collapse
|