1
|
Parsons VA, Vadlamudi S, Voos KM, Rohy AE, Moxley AH, Cannon ME, Rosen JD, Mills CA, Herring LE, Broadaway KA, Lorenzo DN, Mohlke KL. TBC1D30 regulates proinsulin and insulin secretion and is the target of a genomic association signal for proinsulin. Diabetologia 2025; 68:1169-1183. [PMID: 40064677 PMCID: PMC12068983 DOI: 10.1007/s00125-025-06391-w] [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: 10/11/2024] [Accepted: 01/03/2025] [Indexed: 05/13/2025]
Abstract
AIMS/HYPOTHESIS Components of the insulin processing and secretion pathways remain incompletely understood. Here, we examined a genome-wide association study (GWAS) signal for plasma proinsulin levels. Lead GWAS variant rs150781447-T encodes an Arg279Cys substitution in TBC1 domain family member 30 (TBC1D30), but no role for this protein in insulin processing or secretion has been established previously. This study aimed to evaluate whether TBC1D30 drives the GWAS association signal by determining whether TBC1D30 is involved in proinsulin secretion and, if so, to examine the effects of variant alleles and potential mechanisms. METHODS Using CRISPR/Cas9 genome editing to create double-strand breaks and prime editing to install substitutions in INS1 832/13 insulinoma cells, we generated clonal cell lines with altered TBC1D30, as well as homozygous and heterozygous lines carrying the lead GWAS variant. We characterised lines by Sanger sequencing, quantitative PCR and ELISAs to measure glucose-stimulated proinsulin and insulin secretion. We also tested the effects of TBC1D30 knockdown on proinsulin and insulin secretion in human islets. We further assessed TBC1D30's contribution to secretory pathways by examining the effects of altered gene function on intracellular proinsulin and insulin content and insulin localisation, and by identifying potential proteins that interact with TBC1D30 using affinity purification mass spectrometry. RESULTS Compared with mock-edited cells, cell lines with reduced TBC1D30 expression or altered Rab GTPase-activating protein (RabGAP) domain had significantly more secreted proinsulin, 1.8- and 2.6-fold more than controls, respectively. Similarly, cells expressing the variant substitution demonstrated increased proinsulin secretion. Cell lines with a partial deletion of a critical functional domain showed 1.8-fold higher expression of Tbc1d30 and at least 2.0-fold less secreted proinsulin. Cells with altered RabGAP domain sequence also demonstrated, to a lesser extent, changes in secreted insulin levels. TBC1D30 knockdown in human islets resulted in increased insulin secretion with no significant effect on proinsulin secretion. The effects of altered TBC1D30 on mislocalisation of insulin, intracellular proinsulin and insulin content and the identities of interacting proteins are consistent with a role for TBC1D30 in proinsulin and insulin secretion. CONCLUSIONS/INTERPRETATION These findings suggest that effects on TBC1D30 are responsible for the GWAS signal and that TBC1D30 plays a critical role in the secretion of mature insulin.
Collapse
Affiliation(s)
- Victoria A Parsons
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Swarooparani Vadlamudi
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kayleigh M Voos
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Abigail E Rohy
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Anne H Moxley
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Maren E Cannon
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jonathan D Rosen
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Christine A Mills
- UNC Proteomics Core Facility, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Laura E Herring
- UNC Proteomics Core Facility, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - K Alaine Broadaway
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Damaris N Lorenzo
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
2
|
Fossa AJ, Hall AM, Papandonatos GD, Arbuckle TE, Ashley-Martin J, Borghese MM, Bruin J, Chen A, Fisher M, Krzeczkowski JE, Lanphear BP, MacFarlane AJ, Manz KE, Morrison KM, Oulhote Y, Palaniyandi J, Palmert MR, Pennell KD, Vuong AM, Walker DI, Weiler HA, Braun JM. Prenatal PFAS exposures and cardiometabolic health in middle childhood in the MIREC cohort. ENVIRONMENTAL RESEARCH 2025; 274:121330. [PMID: 40057105 DOI: 10.1016/j.envres.2025.121330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 05/04/2025]
Abstract
Studies on prenatal exposure to per- and polyfluoroalkyl substances (PFAS) and cardiometabolic health in childhood have produced inconsistent results. In this study, we evaluated associations between prenatal PFAS exposures, individually and as a mixture, and cardiometabolic outcomes including insulin resistance, beta cell function, blood lipids, blood pressure and central adiposity during middle childhood (7-9 years of age) in a Canadian maternal-child cohort (n = 281). We also explored effect measure modification based on child sex and physical activity. We quantified maternal second trimester plasma concentrations of six PFAS and measured 11 offspring cardiometabolic outcomes at a 7-9-year follow-up. In single-exposure models, ten-fold higher prenatal PFDA (β: -0.82, 95% CI: -1.36, -0.28), PFNA (β: -0.8, 95% CI: -1.41, -0.19), and PFOA (β: -0.69, 95% CI: -1.18, -0.19) concentrations were associated with lower diastolic blood pressure z-scores. This association did not persist when considering PFAS exposures as a mixture using quantile g-computation. Associations between PFAS exposures, individually or as a mixture, and other cardiometabolic outcomes were null. We observed no effect measure modification by child sex or physical activity (p-values for interaction ≥0.2). Our results contradict existing studies that suggest prenatal PFAS exposures are associated with adverse childhood cardiometabolic outcomes. Future studies should consider alternative markers of cardiometabolic health, trajectories in cardiometabolic health throughout childhood, and further explore potentially protective health behaviors.
Collapse
Affiliation(s)
- Alan J Fossa
- Department of Epidemiology, Brown University, Providence, RI, United States
| | - Amber M Hall
- Department of Epidemiology, Brown University, Providence, RI, United States
| | | | - Tye E Arbuckle
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON, Canada
| | | | - Michael M Borghese
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON, Canada
| | - Jenny Bruin
- Department of Biology & Institute of Biochemistry, Carleton University, Ottawa, ON, Canada
| | - Aimin Chen
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Mandy Fisher
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON, Canada
| | | | - Bruce P Lanphear
- Faculty of Health Sciences, Simon Fraser University, Vancouver, BC, Canada
| | - Amanda J MacFarlane
- Nutrition Research Division, Bureau of Nutritional Sciences, Food and Nutrition Directorate, Health Products and Food Branch, Health Canada, Canada
| | - Katherine E Manz
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI, United States
| | - Katherine M Morrison
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Youssef Oulhote
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Jana Palaniyandi
- Department of Biology & Institute of Biochemistry, Carleton University, Ottawa, ON, Canada
| | - Mark R Palmert
- Division of Endocrinology, Hospital for Sick Children: Departments of Pediatrics and Physiology, University of Toronto, Canada
| | - Kurt D Pennell
- School of Engineering, Brown University, Providence, RI, United States
| | - Ann M Vuong
- Department of Epidemiology and Biostatistics, University of Nevada Las Vegas, School of Public Health, Las Vegas, NV, United States
| | - Douglas I Walker
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Hope A Weiler
- Nutrition Research Division, Bureau of Nutritional Sciences, Food and Nutrition Directorate, Health Products and Food Branch, Health Canada, Canada
| | - Joseph M Braun
- Department of Epidemiology, Brown University, Providence, RI, United States.
| |
Collapse
|
3
|
Morales‐Palomo F, Moreno‐Cabañas A, Alvarez‐Jimenez L, Mora‐Gonzalez D, Ortega JF, Mora‐Rodriguez R. Efficacy of morning versus afternoon aerobic exercise training on reducing metabolic syndrome components: A randomized controlled trial. J Physiol 2024; 602:6463-6477. [PMID: 38015017 PMCID: PMC11607890 DOI: 10.1113/jp285366] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 10/16/2023] [Indexed: 11/29/2023] Open
Abstract
A supervised intense aerobic exercise program improves the health of individuals with metabolic syndrome (MetS). However, it is unclear whether the timing of training within the 24 h day would influence those health benefits. The present study aimed to determine the influence of morning vs. afternoon exercise on body composition, cardiometabolic health and components of MetS. One hundred thirty-nine individuals with MetS were block randomized into morning (AMEX; n = 42) or afternoon (PMEX; n = 59) exercise training groups, or a non-training control group (Control; n = 38). Exercise training was comprised of 48 supervised high-intensity interval sessions distributed over 16 weeks. Body composition, cardiorespiratory fitness (assessed byV ̇ O 2 max ${\dot V_{{{\mathrm{O}}_{\mathrm{2}}}{\mathrm{max}}}}$ ), maximal fat oxidation (FOmax), blood pressure and blood metabolites were assessed before and after the intervention. Compared with the non-training Control, both exercise groups improved similarly body composition (-0.7% fat loss; P = 0.002), waist circumference (-2.1 cm; P < 0.001), diastolic blood pressure (-3.8 mmHg; P = 0.004) andV ̇ O 2 max ${\dot V_{{{\mathrm{O}}_{\mathrm{2}}}{\mathrm{max}}}}$ (3.5 mL kg-1 min-1; P < 0.001) with no differences between training groups. AMEX, in comparison with PMEX, reduced systolic blood pressure (-4% vs. -1%; P = 0.019), plasma fasting insulin concentration (-12% vs. -5%; P = 0.001) and insulin resistance (-14% vs. -4%; P = 0.006). Furthermore, MetS Z score was further reduced in the AMEX compared to PMEX (-52% vs. -19%; P = 0.021) after training. In summary, high-intensity aerobic exercise training in the morning in comparison to training in the afternoon is somewhat more efficient at reducing cardiometabolic risk factors (i.e. systolic blood pressure and insulin sensitivity). KEY POINTS: The effect of exercise time of day on health promotion is an area that has gained interest in recent years; however, large-scale, randomized-control studies are scarce. People with metabolic syndrome (MetS) are at risk of developing cardiometabolic diseases and reductions in this risk with exercise training can be precisely gauged using a compound score sensitive to subtle evolution in each MetS component (i.e. Z score). Supervised aerobic exercise for 16 weeks (morning and afternoon), without dietary restriction, improved cardiorespiratory and metabolic fitness, body composition and mean arterial pressure compared to a non-exercise control group. However, training in the morning, without changes in exercise dose or intensity, reduced systolic blood pressure and insulin resistance further compared to when training in the afternoon. Thus, high-intensity aerobic exercise training in the morning is somewhat more efficient in improving the health of individuals with metabolic syndrome.
Collapse
Affiliation(s)
| | - Alfonso Moreno‐Cabañas
- Exercise Physiology Lab at ToledoUniversity of Castilla‐La ManchaToledoSpain
- Centre for Nutrition, Exercise, and MetabolismUniversity of BathBathUK
- Department for HealthUniversity of BathBathUK
| | | | - Diego Mora‐Gonzalez
- Department of Nursing, Physiotherapy and Occupational therapyUniversity of Castilla‐La ManchaToledoSpain
| | - Juan F. Ortega
- Exercise Physiology Lab at ToledoUniversity of Castilla‐La ManchaToledoSpain
| | | |
Collapse
|
4
|
Kosmas CE, Sourlas A, Oikonomakis K, Zoumi EA, Papadimitriou A, Kostara CE. Biomarkers of insulin sensitivity/resistance. J Int Med Res 2024; 52:03000605241285550. [PMCID: PMC11475114 DOI: 10.1177/03000605241285550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/02/2024] [Indexed: 01/03/2025] Open
Abstract
In recent years, remarkable advancements in elucidating the intricate molecular underpinnings of type 2 diabetes mellitus (T2D) have been achieved. Insulin resistance (IR) has been unequivocally acknowledged as the driving pathogenetic mechanism of T2D, preceding disease onset by several years. Nonetheless, diagnostic tools for ascertaining IR are lacking in current clinical practice, representing a critical unmet need; use of the hyperinsulinemic-euglycemic glucose clamp, widely accepted as the gold standard method for evaluating IR at present, is cumbersome in a clinical setting. Thus, the development of well-validated, reliable, and affordable biomarkers of IR has attracted considerable attention from the research community. The biomarkers under investigation can be divided into two major categories: (1) indices or ratios, comprising parameters obtained from a basic or comprehensive metabolic panel and/or derived from anthropometric measurements, and (2) circulating molecules implicated in pathophysiological processes associated with IR. Furthermore, numerous novel biomarkers, including markers of β-cell dysfunction, radiographic quantification of excess visceral adipose tissue, T2D prediction models, certain microRNAs and metabolomic biomarkers, have also provided promising preliminary results. This narrative review aims to present current evidence pertaining to the most notable and exciting biomarkers of IR that are under rigorous evaluation.
Collapse
Affiliation(s)
- Constantine E Kosmas
- Second Department of Cardiology, National & Kapodistrian University of Athens, Athens, Greece
| | | | | | | | | | - Christina E Kostara
- Laboratory of Clinical Chemistry, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| |
Collapse
|
5
|
Curman P, Jebril W, Evans-Molina C, Bachar-Wikstrom E, Larsson H, Cederlöf M, Wikström JD. Hailey-Hailey Disease is Associated with Diabetes: A Population-based Cohort Study, Clinical Cohort Study, and Pedigree Analysis. Acta Derm Venereol 2023; 103:adv10436. [PMID: 38014829 PMCID: PMC10695124 DOI: 10.2340/actadv.v103.10436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 09/27/2023] [Indexed: 11/29/2023] Open
Abstract
Hailey-Hailey disease is a rare hereditary skin disease caused by mutations in the ATP2C1 gene encoding the secretory pathway Ca2+/Mn2+-ATPase 1 (SPCA1) protein. Extracutaneous manifestations of Hailey-Hailey disease are plausible but still largely unknown. The aim of this study was to explore the association between Hailey-Hailey disease and diabetes. A population-based cohort study of 347 individuals with Hailey-Hailey disease was performed to assess the risks of type 1 diabetes and type 2 diabetes, using Swedish nationwide registries. Pedigrees from 2 Swedish families with Hailey-Hailey disease were also investigated: 1 with concurrent type 1 diabetes and HLA-DQ3, the other with type 2 diabetes. Lastly, a clinical cohort with 23 individuals with Hailey-Hailey disease and matched healthy controls was evaluated regarding diabetes. In the register data males with Hailey-Hailey disease had a 70% elevated risk of type 2 diabetes, whereas no excess risk among women could be confirmed. In both pedigrees an unusually high inheritance for diabetes was observed. In the clinical cohort, individuals with Hailey-Hailey disease displayed a metabolic phenotype indicative of type 2 diabetes. Hailey-Hailey disease seems to act as a synergistic risk factor for diabetes. This study indicates, for the first time, an association between Hailey-Hailey disease and diabetes and represents human evidence that SPCA1 and the Golgi apparatus may be implicated in diabetes pathophysiology.
Collapse
Affiliation(s)
- Philip Curman
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden; Dermato-Venereology Clinic, Karolinska University Hospital, Stockholm, Sweden; Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.
| | - William Jebril
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden; Dermato-Venereology Clinic, Karolinska University Hospital, Stockholm, Sweden
| | - Carmella Evans-Molina
- Departments of Anatomy, Cell Biology, and Physiology; Biochemistry and Molecular Biology; Medicine; Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202; The Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202; Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; Roudebush VA Medical Center, Indianapolis, IN 46202, USA
| | - Etty Bachar-Wikstrom
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden; Dermato-Venereology Clinic, Karolinska University Hospital, Stockholm, Sweden; Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Henrik Larsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Martin Cederlöf
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden; School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Jakob D Wikström
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden; Dermato-Venereology Clinic, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
6
|
Marku A, Da Dalt L, Galli A, Dule N, Corsetto P, Rizzo AM, Moregola A, Uboldi P, Bonacina F, Marciani P, Castagna M, Catapano AL, Norata GD, Perego C. Pancreatic PCSK9 controls the organization of the β-cell secretory pathway via LDLR-cholesterol axis. Metabolism 2022; 136:155291. [PMID: 35981632 DOI: 10.1016/j.metabol.2022.155291] [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: 05/23/2022] [Revised: 07/29/2022] [Accepted: 08/11/2022] [Indexed: 11/15/2022]
Abstract
BACKGROUND Cholesterol is central to pancreatic β-cell physiology and alterations of its homeostasis contribute to β-cell dysfunction and diabetes. Proper intracellular cholesterol levels are maintained by different mechanisms including uptake via the low-density lipoprotein receptor (LDLR). In the liver, the proprotein convertase subtilisin/kexin type 9 (PCSK9) routes the LDLR to lysosomes for degradation, thus limiting its recycling to the membrane. PCSK9 is also expressed in the pancreas and loss of function mutations of PCSK9 result in higher plasma glucose levels and increased risk of Type 2 diabetes mellitus. Aim of this study was to investigate whether PCSK9 also impacts β-cells function. METHODS Pancreas-specific Pcsk9 null mice (Pdx1Cre/Pcsk9 fl/fl) were generated and characterized for glucose tolerance, insulin release and islet morphology. Isolated Pcsk9-deficient islets and clonal β-cells (INS1E) were employed to characterize the molecular mechanisms of PCSK9 action. RESULTS Pdx1Cre/Pcsk9 fl/fl mice exhibited normal blood PCSK9 and cholesterol levels but were glucose intolerant and had defective insulin secretion in vivo. Analysis of PCSK9-deficient islets revealed comparable β-cell mass and insulin content but impaired stimulated secretion. Increased proinsulin/insulin ratio, modifications of SNARE proteins expression and decreased stimulated‑calcium dynamics were detected in PCSK9-deficient β-cells. Mechanistically, pancreatic PCSK9 silencing impacts β-cell LDLR expression and cholesterol content, both in vivo and in vitro. The key role of LDLR is confirmed by the demonstration that LDLR downregulation rescued the phenotype. CONCLUSIONS These findings establish pancreatic PCSK9 as a novel critical regulator of the functional maturation of the β-cell secretory pathway, via modulation of cholesterol homeostasis.
Collapse
Affiliation(s)
- Algerta Marku
- Dept of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20134 Milan, Italy
| | - Lorenzo Da Dalt
- Dept of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20134 Milan, Italy
| | - Alessandra Galli
- Dept of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20134 Milan, Italy
| | - Nevia Dule
- Dept of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20134 Milan, Italy
| | - Paola Corsetto
- Dept of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20134 Milan, Italy
| | - Angela Maria Rizzo
- Dept of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20134 Milan, Italy
| | - Annalisa Moregola
- Dept of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20134 Milan, Italy
| | - Patrizia Uboldi
- Dept of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20134 Milan, Italy
| | - Fabrizia Bonacina
- Dept of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20134 Milan, Italy
| | - Paola Marciani
- Dept of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20134 Milan, Italy
| | - Michela Castagna
- Dept of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20134 Milan, Italy
| | - Alberico Luigi Catapano
- Dept of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20134 Milan, Italy; IRCCS Multimedica Hospital, Sesto San Giovanni, 20099 Milan, Italy
| | - Giuseppe Danilo Norata
- Dept of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20134 Milan, Italy; Centro SISA per lo studio dell'Aterosclerosi, Ospedale Bassini, 20092 Cinisello Balsamo, Italy.
| | - Carla Perego
- Dept of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20134 Milan, Italy.
| |
Collapse
|
7
|
Fasting Proinsulin Independently Predicts Incident Type 2 Diabetes in the General Population. J Pers Med 2022; 12:jpm12071131. [PMID: 35887628 PMCID: PMC9323856 DOI: 10.3390/jpm12071131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 06/29/2022] [Accepted: 07/05/2022] [Indexed: 11/17/2022] Open
Abstract
Fasting proinsulin levels may serve as a marker of β-cell dysfunction and predict type 2 diabetes (T2D) development. Kidneys have been found to be a major site for the degradation of proinsulin. We aimed to evaluate the predictive value of proinsulin for the risk of incident T2D added to a base model of clinical predictors and examined potential effect modification by variables related to kidney function. Proinsulin was measured in plasma with U-PLEX platform using ELISA immunoassay. We included 5001 participants without T2D at baseline and during a median follow up of 7.2 years; 271 participants developed T2D. Higher levels of proinsulin were associated with increased risk of T2D independent of glucose, insulin, C-peptide, and other clinical factors (hazard ratio (HR): 1.28; per 1 SD increase 95% confidence interval (CI): 1.08–1.52). Harrell’s C-index for the Framingham offspring risk score was improved with the addition of proinsulin (p = 0.019). Furthermore, we found effect modification by hypertension (p = 0.019), eGFR (p = 0.020) and urinary albumin excretion (p = 0.034), consistent with an association only present in participants with hypertension or kidney dysfunction. Higher fasting proinsulin level is an independent predictor of incident T2D in the general population, particularly in participants with hypertension or kidney dysfunction.
Collapse
|
8
|
Kahn SE, Chen YC, Esser N, Taylor AJ, van Raalte DH, Zraika S, Verchere CB. The β Cell in Diabetes: Integrating Biomarkers With Functional Measures. Endocr Rev 2021; 42:528-583. [PMID: 34180979 PMCID: PMC9115372 DOI: 10.1210/endrev/bnab021] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Indexed: 02/08/2023]
Abstract
The pathogenesis of hyperglycemia observed in most forms of diabetes is intimately tied to the islet β cell. Impairments in propeptide processing and secretory function, along with the loss of these vital cells, is demonstrable not only in those in whom the diagnosis is established but typically also in individuals who are at increased risk of developing the disease. Biomarkers are used to inform on the state of a biological process, pathological condition, or response to an intervention and are increasingly being used for predicting, diagnosing, and prognosticating disease. They are also proving to be of use in the different forms of diabetes in both research and clinical settings. This review focuses on the β cell, addressing the potential utility of genetic markers, circulating molecules, immune cell phenotyping, and imaging approaches as biomarkers of cellular function and loss of this critical cell. Further, we consider how these biomarkers complement the more long-established, dynamic, and often complex measurements of β-cell secretory function that themselves could be considered biomarkers.
Collapse
Affiliation(s)
- Steven E Kahn
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, VA Puget Sound Health Care System and University of Washington, Seattle, 98108 WA, USA
| | - Yi-Chun Chen
- BC Children's Hospital Research Institute and Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| | - Nathalie Esser
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, VA Puget Sound Health Care System and University of Washington, Seattle, 98108 WA, USA
| | - Austin J Taylor
- BC Children's Hospital Research Institute and Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| | - Daniël H van Raalte
- Department of Internal Medicine, Amsterdam University Medical Center (UMC), Vrije Universiteit (VU) University Medical Center, 1007 MB Amsterdam, The Netherlands.,Department of Experimental Vascular Medicine, Amsterdam University Medical Center (UMC), Academic Medical Center, 1007 MB Amsterdam, The Netherlands
| | - Sakeneh Zraika
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, VA Puget Sound Health Care System and University of Washington, Seattle, 98108 WA, USA
| | - C Bruce Verchere
- BC Children's Hospital Research Institute and Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| |
Collapse
|
9
|
Shigeno R, Horie I, Miwa M, Ito A, Haraguchi A, Natsuda S, Akazawa S, Nagata A, Hasegawa Y, Miura S, Miura K, Kawakami A, Abiru N. Bihormonal dysregulation of insulin and glucagon contributes to glucose intolerance development at one year post-delivery in women with gestational diabetes: a prospective cohort study using an early postpartum 75-g glucose tolerance test. Endocr J 2021; 68:919-931. [PMID: 33827994 DOI: 10.1507/endocrj.ej20-0795] [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] [Indexed: 11/23/2022] Open
Abstract
Gestational diabetes mellitus (GDM) is known to be a significant risk factor for the future development of type 2 diabetes. Here, we investigated whether a precise evaluation of β- and α-cell functions helps to identify women at high risk of developing glucose intolerance after GDM. Fifty-six women with GDM underwent a 75-g oral glucose tolerance test (OGTT) at early (6-12 weeks) postpartum. We measured their concentrations of glucose, insulin, proinsulin and glucagon at fasting and 30, 60 and 120 min. At 1-year post-delivery, we classified the women into a normal glucose tolerance (NGT) group or an impaired glucose tolerance (IGT)/diabetes mellitus (DM) group. Forty-three of the 56 women completed the study. At 1-year post-delivery, 17 women had developed IGT/DM and 26 women showed NGT. In the early-postpartum OGTTs, the IGT/DM group showed a lower insulinogenic index, a less glucagon suppression evaluated by the change from fasting to 30 min (ΔGlucagon 30 min), and a higher glucagon-to-insulin ratio at 30 min compared to the NGT group. There were no significant between-group differences in proinsulin levels or proinsulin-to-insulin ratios. Insulinogenic index <0.6 and ΔGlucagon 30 min >0 pg/mL were identified as predictors for the development of IGT/DM after GDM, independent of age, body mass index, and lactation intensity. These results suggest that the bihormonal disorder of insulin and glucagon causes the postpartum development of glucose intolerance. The measurement of plasma insulin and glucagon during the initial OGTT at early postpartum period can help to make optimal decisions regarding the postpartum management of women with GDM.
Collapse
Affiliation(s)
- Riyoko Shigeno
- Department of Endocrinology and Metabolism, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Ichiro Horie
- Department of Endocrinology and Metabolism, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Masaki Miwa
- Department of Endocrinology and Metabolism, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Ayako Ito
- Department of Endocrinology and Metabolism, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Ai Haraguchi
- Department of Endocrinology and Metabolism, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Shoko Natsuda
- Department of Endocrinology and Metabolism, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Satoru Akazawa
- Department of Endocrinology and Metabolism, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Ai Nagata
- Department of Obstetrics and Gynecology, Nagasaki University Hospital, Nagasaki 852-8501, Japan
| | - Yuri Hasegawa
- Department of Obstetrics and Gynecology, Nagasaki University Hospital, Nagasaki 852-8501, Japan
| | - Shoko Miura
- Department of Obstetrics and Gynecology, Nagasaki University Hospital, Nagasaki 852-8501, Japan
| | - Kiyonori Miura
- Department of Obstetrics and Gynecology, Nagasaki University Hospital, Nagasaki 852-8501, Japan
| | - Atsushi Kawakami
- Department of Endocrinology and Metabolism, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Norio Abiru
- Department of Endocrinology and Metabolism, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| |
Collapse
|
10
|
Germanos M, Gao A, Taper M, Yau B, Kebede MA. Inside the Insulin Secretory Granule. Metabolites 2021; 11:metabo11080515. [PMID: 34436456 PMCID: PMC8401130 DOI: 10.3390/metabo11080515] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/03/2021] [Accepted: 08/03/2021] [Indexed: 12/19/2022] Open
Abstract
The pancreatic β-cell is purpose-built for the production and secretion of insulin, the only hormone that can remove glucose from the bloodstream. Insulin is kept inside miniature membrane-bound storage compartments known as secretory granules (SGs), and these specialized organelles can readily fuse with the plasma membrane upon cellular stimulation to release insulin. Insulin is synthesized in the endoplasmic reticulum (ER) as a biologically inactive precursor, proinsulin, along with several other proteins that will also become members of the insulin SG. Their coordinated synthesis enables synchronized transit through the ER and Golgi apparatus for congregation at the trans-Golgi network, the initiating site of SG biogenesis. Here, proinsulin and its constituents enter the SG where conditions are optimized for proinsulin processing into insulin and subsequent insulin storage. A healthy β-cell is continually generating SGs to supply insulin in vast excess to what is secreted. Conversely, in type 2 diabetes (T2D), the inability of failing β-cells to secrete may be due to the limited biosynthesis of new insulin. Factors that drive the formation and maturation of SGs and thus the production of insulin are therefore critical for systemic glucose control. Here, we detail the formative hours of the insulin SG from the luminal perspective. We do this by mapping the journey of individual members of the SG as they contribute to its genesis.
Collapse
|
11
|
Quan H, Fang T, Lin L, Lin L, Ou Q, Zhang H, Chen K, Zhou Z. The Impact of Different Insulin-Related Measures on the Risk of Prediabetes Among the Chinese Han Population. Diabetes Ther 2021; 12:2195-2206. [PMID: 34236576 PMCID: PMC8342746 DOI: 10.1007/s13300-021-01102-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/15/2021] [Indexed: 11/26/2022] Open
Abstract
INTRODUCTION Diabetes mellitus (DM) has a serious impact on people's lives in the world. Interventions that affect risk factors for prediabetes can prevent and reduce diabetes occurrence. Proinsulin (PI), true insulin (TI), and proinsulin to insulin ratio (PI/TI) are risk factors for diabetes. The roles of these indicators in prediabetes are unclear. This study aimed to evaluate the impact of PI, TI, PI/TI, 2-h proinsulin (2hPI), 2-h true insulin (2hTI), and 2hPI/2hTI on the risk of prediabetes among the Chinese Han population. METHODS This cross-sectional study recruited 1688 subjects including 718 prediabetes cases and 970 non-prediabetes controls from Hainan Affiliated Hospital of Hainan Medical University. The cases involved 292 men and 426 women. The controls involved 324 men and 646 women. The mean age was 53.62 ± 12.43 years in the prediabetes group and 44.24 ± 12.87 years in the non-prediabetes group. RESULTS Our results showed that PI, TI, PI/TI, 2hPI, 2hTI, and 2hPI/2hTI were significantly correlated with prediabetes (all p < 0.05). Logistic regression analysis revealed that PI (OR 1.022, 95% CI 1.014-1.031, p = 0.00011), TI (OR 1.005, 95% CI 1.003-1.007, p = 0.00012), PI/TI (OR 1.517, 95% CI 1.080-2.131, p = 0.016), and 2hTI (OR 1.000, 95% CI 1.000-1.001, p = 0.002) were significantly associated with an increased risk of prediabetes. Receiver operating characteristic curve (ROC) analysis indicated that the area under the ROC curve (AUC) of the combination (PI + TI + PI/TI + 2hPI + 2hTI + 2hPI/2hTI) in diagnosing prediabetes was 0.627, which was larger than the diagnostic value of HOMA-IR (AUC 0.614) and HOMA-β (AUC 0.387). CONCLUSIONS Our study showed that PI, TI, PI/TI, and 2hTI could significantly enhance the risk of prediabetes in the Chinese Han population, which suggested that PI, TI, PI/TI, and 2hTI might be available risk factors for prediabetes.
Collapse
Affiliation(s)
- Huibiao Quan
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, No. 139, Renmin Middle Road, Furong District, Changsha, 410011, Hunan Province, China
- Department of Endocrinology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No.19, Xiuhua Road, Haikou, 570311, Hainan, China
| | - Tuanyu Fang
- Department of Endocrinology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No.19, Xiuhua Road, Haikou, 570311, Hainan, China
| | - Leweihua Lin
- Department of Endocrinology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No.19, Xiuhua Road, Haikou, 570311, Hainan, China
| | - Lu Lin
- Department of Endocrinology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No.19, Xiuhua Road, Haikou, 570311, Hainan, China
| | - Qianying Ou
- Department of Endocrinology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No.19, Xiuhua Road, Haikou, 570311, Hainan, China
| | - Huachuan Zhang
- Department of Endocrinology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No.19, Xiuhua Road, Haikou, 570311, Hainan, China
| | - Kaining Chen
- Department of Endocrinology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No.19, Xiuhua Road, Haikou, 570311, Hainan, China
| | - Zhiguang Zhou
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, No. 139, Renmin Middle Road, Furong District, Changsha, 410011, Hunan Province, China.
| |
Collapse
|
12
|
Ramzy A, Kieffer TJ. Altered islet prohormone processing: A cause or consequence of diabetes? Physiol Rev 2021; 102:155-208. [PMID: 34280055 DOI: 10.1152/physrev.00008.2021] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Peptide hormones are first produced as larger precursor prohormones that require endoproteolytic cleavage to liberate the mature hormones. A structurally conserved but functionally distinct family of nine prohormone convertase enzymes (PCs) are responsible for cleavage of protein precursors of which PC1/3 and PC2 are known to be exclusive to neuroendocrine cells and responsible for prohormone cleavage. Differential expression of PCs within tissues define prohormone processing; whereas glucagon is the major product liberated from proglucagon via PC2 in pancreatic α-cells, proglucagon is preferentially processed by PC1/3 in intestinal L cells to produce glucagon-like peptides 1 and 2 (GLP-1, GLP-2). Beyond our understanding of processing of islet prohormones in healthy islets, there is convincing evidence that proinsulin, proIAPP, and proglucagon processing is altered during prediabetes and diabetes. There is predictive value of elevated circulating proinsulin or proinsulin : C-peptide ratio for progression to type 2 diabetes and elevated proinsulin or proinsulin : C-peptide is predictive for development of type 1 diabetes in at risk groups. After onset of diabetes, patients have elevated circulating proinsulin and proIAPP and proinsulin may be an autoantigen in type 1 diabetes. Further, preclinical studies reveal that α-cells have altered proglucagon processing during diabetes leading to increased GLP-1 production. We conclude that despite strong associative data, current evidence is inconclusive on the potential causal role of impaired prohormone processing in diabetes, and suggest that future work should focus on resolving the question of whether altered prohormone processing is a causal driver or merely a consequence of diabetes pathology.
Collapse
Affiliation(s)
- Adam Ramzy
- Laboratory of Molecular and Cellular Medicine, Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Timothy J Kieffer
- Laboratory of Molecular and Cellular Medicine, Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, Canada.,School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
13
|
Isolation and Proteomics of the Insulin Secretory Granule. Metabolites 2021; 11:metabo11050288. [PMID: 33946444 PMCID: PMC8147143 DOI: 10.3390/metabo11050288] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 04/28/2021] [Accepted: 04/28/2021] [Indexed: 12/21/2022] Open
Abstract
Insulin, a vital hormone for glucose homeostasis is produced by pancreatic beta-cells and when secreted, stimulates the uptake and storage of glucose from the blood. In the pancreas, insulin is stored in vesicles termed insulin secretory granules (ISGs). In Type 2 diabetes (T2D), defects in insulin action results in peripheral insulin resistance and beta-cell compensation, ultimately leading to dysfunctional ISG production and secretion. ISGs are functionally dynamic and many proteins present either on the membrane or in the lumen of the ISG may modulate and affect different stages of ISG trafficking and secretion. Previously, studies have identified few ISG proteins and more recently, proteomics analyses of purified ISGs have uncovered potential novel ISG proteins. This review summarizes the proteins identified in the current ISG proteomes from rat insulinoma INS-1 and INS-1E cell lines. Here, we also discuss techniques of ISG isolation and purification, its challenges and potential future directions.
Collapse
|
14
|
Quan H, Fang T, Lin L, Lin L, Ou Q, Zhang H, Chen K, Zhou Z. The correlation between proinsulin, true insulin, proinsulin: True insulin ratio, 25(OH) D3, waist circumference and risk of prediabetes in Hainan Han adults. PLoS One 2020; 15:e0238095. [PMID: 32881889 PMCID: PMC7470272 DOI: 10.1371/journal.pone.0238095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 08/10/2020] [Indexed: 11/18/2022] Open
Abstract
Purpose Diabetes mellitus is a kind of highly prevalent chronic disease in the world. The intervention measures on the risk factors of prediabetes contribute to control and reduce the occurrence of diabetes. This study aimed to investigate the correlation between proinsulin (PI), true insulin (TI), PI/TI, 25(OH) D3, waist circumference (WC), and risk of prediabetes. Methods In this cross-sectional study, 1662 subjects including 615 prediabetes and 1047 non-prediabetes were recruited. Spearman’s correlation analysis was used to explore the association of PI, TI, PI/TI, 25(OH) D3, and waist circumference with prediabetes. Odds ratios (OR) and 95% confidence intervals (CI) were calculated by logistic regression. Receiver-Operator Characteristic (ROC) curve was used to evaluate the risk of prediabetes. Results Our study showed that FPI, 2hPI, FTI, 2hTI, FPI/FTI, and WC could enhance the risk of prediabetes (OR 1.034; OR 1.007; OR 1.005; OR 1.002; OR 3.577, OR 1.053, respectively; all p< 0.001). Stratified analyses indicated that FPI/FTI associated with an increased risk of prediabetes in men (OR 2.080, p = 0.042). FTI have a weak association with prediabetes risk in men and women (OR 0.987, p = 0.001; OR 0.994, p = 0.004, respectively). 2hPI could decrease prediabetes in women (OR 0.995, p = 0.037). Interesting, the sensitivity (86.0%) and AUC (0.942, p< 0.001) of combination (FPI+FTI+2hPI+2hTI+25(OH) D3+WC) were higher than the diagnostic value of these alone diagnoses. The optimal cutoff point of FPI, FTI, 2hPI, 2hTI, 25(OH) D3, and WC for indicating prediabetes were 15.5 mU/l, 66.5 mU/l, 71.5 mU/l, 460.5 mU/l, 35.5 ng/ml, and 80.5 cm, respectively. What’s more, the combination (FPI+FTI+2hPI+2hTI+25(OH) D3+WC) significantly improved the diagnostic value beyond the alone diagnoses of prediabetes in men and women (AUC 0.771; AUC 0.760, respectively). Conclusion The FPI, 2hPI, FTI, 2hTI, FPI/FTI, and WC significantly associated with an increased risk of prediabetes. The combination of FPI, FTI, 2hPI, 2hTI, 25(OH) D3, and WC might be used as diagnostic indicators for prediabetes.
Collapse
Affiliation(s)
- Huibiao Quan
- Department of Metabolism & Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Endocrinology, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Tuanyu Fang
- Department of Endocrinology, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Leweihua Lin
- Department of Endocrinology, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Lu Lin
- Department of Endocrinology, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Qianying Ou
- Department of Endocrinology, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Huachuan Zhang
- Department of Endocrinology, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Kaining Chen
- Department of Endocrinology, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Zhiguang Zhou
- Department of Metabolism & Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- * E-mail:
| |
Collapse
|
15
|
Li T, Quan H, Zhang H, Lin L, Lin L, Ou Q, Chen K. Subgroup analysis of proinsulin and insulin levels reveals novel correlations to metabolic indicators of type 2 diabetes. Aging (Albany NY) 2020; 12:10715-10735. [PMID: 32532930 DOI: 10.18632/aging.103289] [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: 12/11/2019] [Accepted: 04/27/2020] [Indexed: 11/25/2022]
Abstract
Proinsulin, insulin and proinsulin/insulin (P/I) ratio have been reported to be correlated with fasting plasma glucose (FPG) and Hemoglobin A1c (HbA1c) in whole population study therefore sensitive predictors of T2D progression. However, by analyzing data collected from 2018-2019 from a cohort of 1579 East Asian individuals from Hainan Province of China, we find that the associations of proinsulin, insulin and P/I ratio with diabetic indicators have distinct, sometimes opposite regression patterns in normal, prediabetic and diabetic subgroups. The strength of the associations are generally weak in normal and prediabetic groups, and only moderate in diabetic group between postprandial proinsulin and HbA1c, between postprandial insulin and FPG or HbA1c, and between postprandial P/I ratio and FPG or HbA1c. Receiver operating characteristic (ROC) curve analysis shows these parameters are weaker than age in predicting diabetes development, with P/I ratio being the weakest. Proinsulin and insulin levels are tightly associated with insulin sensitivity across all subgroups, as measured by Matsuda index. Together, our results suggest that proinsulin, insulin or P/I ratio are weak predictors of diabetes development in the whole population, urging the need for stratifying strategies and novel perspectives in evaluating and predicting hyperglycemia progression.
Collapse
Affiliation(s)
- Tangying Li
- Department of Health Care Centre, Hainan General Hospital, Haikou 570311, Hainan, China
| | - Huibiao Quan
- Department of Endocrinology, Hainan General Hospital, Haikou 570311, Hainan, China
| | - Huachuan Zhang
- Department of Endocrinology Laboratory, Hainan General Hospital, Haikou 570311, Hainan, China
| | - Leweihua Lin
- Department of Endocrinology, Hainan General Hospital, Haikou 570311, Hainan, China
| | - Lu Lin
- Department of Endocrinology, Hainan General Hospital, Haikou 570311, Hainan, China
| | - Qianying Ou
- Department of Endocrinology, Hainan General Hospital, Haikou 570311, Hainan, China
| | - Kaining Chen
- Department of Endocrinology, Hainan General Hospital, Haikou 570311, Hainan, China
| |
Collapse
|
16
|
Abstract
The environment within the Endoplasmic Reticulum (ER) influences Insulin biogenesis. In particular, ER stress may contribute to the development of Type 2 Diabetes (T2D) and Cystic Fibrosis Related Diabetes (CFRD), where evidence of impaired Insulin processing, including elevated secreted Proinsulin/Insulin ratios, are observed. Our group has established the role of a novel ER chaperone ERp29 (ER protein of 29 kDa) in the biogenesis of the Epithelial Sodium Channel, ENaC. The biogenesis of Insulin and ENaC share may key features, including their potential association with COP II machinery, their cleavage into a more active form in the Golgi or later compartments, and their ability to bypass such cleavage and remain in a less active form. Given these similarities we hypothesized that ERp29 is a critical factor in promoting the efficient conversion of Proinsulin to Insulin. Here, we confirmed that Proinsulin associates with the COP II vesicle cargo recognition component, Sec24D. When Sec24D expression was decreased, we observed a corresponding decrease in whole cell Proinsulin levels. In addition, we found that Sec24D associates with ERp29 in co-precipitation experiments and that ERp29 associates with Proinsulin in co-precipitation experiments. When ERp29 was overexpressed, a corresponding increase in whole cell Proinsulin levels was observed, while depletion of ERp29 decreased whole cell Proinsulin levels. Together, these data suggest a potential role for ERp29 in regulating Insulin biosynthesis, perhaps in promoting the exit of Proinsulin from the ER via Sec24D/COPII vesicles.
Collapse
|
17
|
Then C, Gar C, Thorand B, Huth C, Then H, Meisinger C, Heier M, Peters A, Koenig W, Rathmann W, Lechner A, Seissler J. Proinsulin to insulin ratio is associated with incident type 2 diabetes but not with vascular complications in the KORA F4/FF4 study. BMJ Open Diabetes Res Care 2020; 8:8/1/e001425. [PMID: 32423965 PMCID: PMC7245418 DOI: 10.1136/bmjdrc-2020-001425] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/20/2020] [Accepted: 04/26/2020] [Indexed: 01/14/2023] Open
Abstract
INTRODUCTION We investigated the association of the proinsulin to insulin ratio (PIR) with prevalent and incident type 2 diabetes (T2D), components of the metabolic syndrome, and renal and cardiovascular outcomes in the population-based Cooperative Health Research in the Region of Augsburg (KORA) F4 study (2006-2008)/FF4 study (2013-2014). RESEARCH DESIGN AND METHODS The analyses included 1514 participants of the KORA F4 study at baseline and 1132 participants of the KORA FF4 study after a median follow-up time of 6.6 years. All-cause and cardiovascular mortality as well as cardiovascular events were analyzed after a median time of 9.1 and 8.6 years, respectively. The association of PIR with T2D, renal and cardiovascular characteristics and mortality were assessed using logistic regression models. Linear regression analyses were used to assess the association of PIR with components of the metabolic syndrome. RESULTS After adjustment for sex, age, body mass index (BMI), and physical activity, PIR was associated with prevalent (OR: 2.24; 95% CI 1.81 to 2.77; p<0.001) and incident T2D (OR: 1.66; 95% CI 1.26 to 2.17; p<0.001). PIR was associated with fasting glucose (β per SD: 0.11±0.02; p<0.001) and HbA1c (β: 0.21±0.02; p<0.001). However, PIR was not positively associated with other components of the metabolic syndrome and was even inversely associated with waist circumference (β: -0.22±0.03; p<0.001), BMI (β: -0.11±0.03; p<0.001) and homeostatic model assessment of insulin resistance (β: -0.22±0.02; p<0.001). PIR was not significantly associated with the intima-media thickness (IMT), decline of kidney function, incident albuminuria, myocardial infarction, stroke, cardiovascular or all-cause mortality. CONCLUSIONS In the KORA F4/FF4 cohort, PIR was positively associated with prevalent and incident T2D, but inversely associated with waist circumference, BMI and insulin resistance, suggesting that PIR might serve as a biomarker for T2D risk independently of the metabolic syndrome, but not for microvascular or macrovascular complications.
Collapse
Affiliation(s)
- Cornelia Then
- Medizinische Klinik und Poliklinik IV, LMU Klinikum der Universität München, Munich, Germany
| | - Christina Gar
- Medizinische Klinik und Poliklinik IV, LMU Klinikum der Universität München, Munich, Germany
- Clinical Cooperation Group Diabetes, Ludwig-Maximilians-Universität München and Helmholtz Zentrum München, Munich, Germany
| | - Barbara Thorand
- Institute of Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
- Deutsches Zentrum für Diabetesforschung (DZD), German Center for Diabetes Research, München-Neuherberg, Germany
| | - Cornelia Huth
- Institute of Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
- Deutsches Zentrum für Diabetesforschung (DZD), German Center for Diabetes Research, München-Neuherberg, Germany
| | - Holger Then
- Department of Mathematics, Freie Waldorfschule Augsburg, Augsburg, Germany
| | - Christa Meisinger
- Institute of Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
- Chair of Epidemiology at UNIKA-T Augsburg, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Margit Heier
- Institute of Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
- KORA Study Centre, University Hospital Augsburg, Augsburg, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
- Deutsches Zentrum für Diabetesforschung (DZD), German Center for Diabetes Research, München-Neuherberg, Germany
| | - Wolfgang Koenig
- Institute of Epidemiology and Medical Biometry, Ulm University, Ulm, Germany
- Technische Universität München, Deutsches Herzzentrum München, München, Germany
| | - Wolfgang Rathmann
- German Diabetes Center, Institute of Biometrics and Epidemiology, Leibniz Institute at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Andreas Lechner
- Medizinische Klinik und Poliklinik IV, LMU Klinikum der Universität München, Munich, Germany
- Clinical Cooperation Group Diabetes, Ludwig-Maximilians-Universität München and Helmholtz Zentrum München, Munich, Germany
| | - Jochen Seissler
- Medizinische Klinik und Poliklinik IV, LMU Klinikum der Universität München, Munich, Germany
- Clinical Cooperation Group Diabetes, Ludwig-Maximilians-Universität München and Helmholtz Zentrum München, Munich, Germany
| |
Collapse
|
18
|
Yang Y, Wang M, Tong J, Dong Z, Deng M, Ren X, Li H, Yang J, Meng Z, Sun J, He Q, Liu M. Impaired Glucose-Stimulated Proinsulin Secretion Is an Early Marker of β-Cell Impairment Before Prediabetes Stage. J Clin Endocrinol Metab 2019; 104:4341-4346. [PMID: 31074785 DOI: 10.1210/jc.2019-00549] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 05/06/2019] [Indexed: 01/09/2023]
Abstract
CONTEXT Evidence indicates that there is substantial impairment/loss of β-cell function/mass even before prediabetes. Elevated plasma proinsulin is a sign of β-cell dysfunction in patients with diabetes/prediabetes. However, the dynamic changes of glucose stimulated proinsulin secretion (GSPS) among nondiabetic individuals remain obscure. OBJECTIVE To examine GSPS and glucose-stimulated insulin secretion (GSIS) among individuals with normal glucose tolerance (NGT) and impaired glucose tolerance (IGT) and to evaluate whether impaired GSPS is an early biomarker of β-cell impairment in individuals with NGT who have subthreshold postprandial plasma glucose (PPG). DESIGN AND PARTICIPANTS We evaluated GSPS and GSIS in 116 Chinese adults without diabetes (mean age ± SD, 33.31 ± 9.10 years; mean BMI, 25.24 ± 4.20 kg/m2) with fasting plasma glucose (FPG) < 5.6 mmol/L. Based on 2hPPG, the participants were divided into three groups: NGT1 (2hPPG < 6.67 mmol/L), NGT2 (6.67 ≤ 2hPPG < 7.78 mmol/L), and IGT (7.78 ≤ 2hPPG<11.1 mmol/L). We analyzed the association of GSIS and GSPS with commonly used indexes of β-cell function, insulin resistance and family history of diabetes. RESULTS Although not diagnosed with prediabetes, the individuals with NGT2 have clinical characteristics and high diabetes risk factors similar to those of the IGT group. However, unlike individuals with IGT, NGT2 participants did not exhibit a delayed GSIS. Instead, GSPS was impaired in NGT2 groups but not in NGT1 group. CONCLUSIONS This study suggests that impaired GSPS, but not impaired GSIS, may serve as an early biomarker to identify a subpopulation of NGT with a high risk of diabetes.
Collapse
Affiliation(s)
- Ying Yang
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Min Wang
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Jingzhi Tong
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Zuoliang Dong
- Department of Medical Laboratory, Tianjin Medical University General Hospital, Tianjin, China
| | - Min Deng
- Department of Physical Examination, Tianjin Electric Power Hospital, Tianjin, China
| | - Xiaojun Ren
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Hui Li
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Jing Yang
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhaowei Meng
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Jinhong Sun
- Department of Health Management, Tianjin Medical University General Hospital, Tianjin, China
| | - Qing He
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Ming Liu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
19
|
Effects of coconut oil on glycemia, inflammation, and urogenital microbial parameters in female Ossabaw mini-pigs. PLoS One 2017; 12:e0179542. [PMID: 28704429 PMCID: PMC5509134 DOI: 10.1371/journal.pone.0179542] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 05/31/2017] [Indexed: 02/07/2023] Open
Abstract
Forty percent of American women are obese and at risk for type II diabetes, impaired immune function, and altered microbiome diversity, thus impacting overall health. We investigated whether obesity induced by an excess calorie, high fat diet containing hydrogenated fats, fructose, and coconut oil (HFD) altered glucose homeostasis, peripheral immunity, and urogenital microbial dynamics. We hypothesized that HFD would cause hyperglycemia, increase peripheral inflammation, and alter urogenital microbiota to favor bacterial taxonomy associated with inflammation. We utilized female Ossabaw mini-pigs to model a ‘thrifty’ metabolic phenotype associated with increased white adipose tissue mass. Pigs were fed HFD (~4570 kcal/pig/day) or lean (~2000 kcal/pig/day) diet for a total of 9 estrous cycles (~6 months). To determine the effect of cycle stage on cytokines and the microbiome, animals had samples collected during cycles 7 and 9 on certain days of the cycle: D1, 4, 8, 12, 16, 18. Vaginal swabs or cervical flushes assessed urogenital microbiota. Systemic fatty acids, insulin, glucose, and cytokines were analyzed. Pig weights and morphometric measurements were taken weekly. Obese pigs had increased body weight, length, heart and belly girth but similar glucose concentrations. Obese pigs had decreased cytokine levels (IL-1β, TNF-α, IL-4, IL-10), arachidonic acid and plasma insulin, but increased levels of vaccenic acid. Obese pigs had greater urogenital bacterial diversity, including several taxa known for anti-inflammatory properties. Overall, induction of obesity did not induce inflammation but shifted the microbial communities within the urogenital tract to an anti-inflammatory phenotype. We postulate that the coconut oil in the HFD oil may have supported normal glucose homeostasis and modulated the immune response, possibly through regulation of microbial community dynamics and fatty acid metabolism. This animal model holds promise for the study of how different types of obesity and high fat diets may affect metabolism, immune phenotype, and microbial dynamics.
Collapse
|
20
|
Courtade JA, Klimek-Abercrombie AM, Chen YC, Patel N, Lu PYT, Speake C, Orban PC, Najafian B, Meneilly G, Greenbaum CJ, Warnock GL, Panagiotopoulos C, Verchere CB. Measurement of Pro-Islet Amyloid Polypeptide (1-48) in Diabetes and Islet Transplants. J Clin Endocrinol Metab 2017; 102:2595-2603. [PMID: 28368485 DOI: 10.1210/jc.2016-2773] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 03/20/2017] [Indexed: 12/21/2022]
Abstract
CONTEXT Islet amyloid is a feature of β-cell failure in type 2 diabetes (T2D) and type 1 diabetes (T1D) recipients of islet transplants. Islet amyloid contains islet amyloid polypeptide (IAPP; amylin), a circulating peptide that is produced in β cells by processing of its precursor, proIAPP1-67, via an intermediate form, proIAPP1-48. Elevated proinsulin to C-peptide ratios in the plasma of persons with diabetes suggest defects in β-cell prohormone processing. OBJECTIVE Determine whether plasma levels of precursor forms of IAPP are elevated in diabetes. DESIGN, SETTING, AND PATIENTS We developed an immunoassay to detect proIAPP1-48 in human plasma, and we determined the ratio of proIAPP1-48 to mature IAPP in subjects with T1D, T2D, recipients of islet transplants, and healthy controls. RESULTS The proIAPP1-48 immunoassay had a limit of detection of 0.18 ± 0.06 pM and cross-reactivity with intact proIAPP1-67 <15%. Healthy individuals had plasma concentrations of proIAPP1-48 immunoreactivity of 1.5 ± 0.2 pM and a proIAPP1-48 to total IAPP ratio of 0.28 ± 0.03. Plasma concentrations of proIAPP1-48 immunoreactivity were not significantly different in subjects with T2D but were markedly increased in T1D recipients of islet transplants. Children and adults with T1D had reduced mature IAPP levels relative to age-matched controls but an elevated ratio of proIAPP1-48 to total IAPP. CONCLUSION The β cells in T1D and islet transplants have impaired processing of the proIAPP1-48 intermediate. The ratio of proIAPP1-48-to-IAPP immunoreactivity may have value as a biomarker of β-cell stress and dysfunction.
Collapse
Affiliation(s)
- Jaques A Courtade
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
- Research Institute, BC Children's Hospital, Vancouver, British Columbia V5Z 4H4, Canada
| | - Agnieszka M Klimek-Abercrombie
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
- Research Institute, BC Children's Hospital, Vancouver, British Columbia V5Z 4H4, Canada
| | - Yi-Chun Chen
- Research Institute, BC Children's Hospital, Vancouver, British Columbia V5Z 4H4, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - Nirja Patel
- American Laboratory Products Company, Salem, New Hampshire 03079
| | - Phoebe Y T Lu
- Research Institute, BC Children's Hospital, Vancouver, British Columbia V5Z 4H4, Canada
- Department of Genetics, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - Cate Speake
- Diabetes Clinical Research Program, Benaroya Research Institute, Seattle, Washington 98101
| | - Paul C Orban
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
- Research Institute, BC Children's Hospital, Vancouver, British Columbia V5Z 4H4, Canada
| | - Behzad Najafian
- Department of Pathology, University of Washington, Seattle, Washington 98195
| | - Graydon Meneilly
- Department of Medicine, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - Carla J Greenbaum
- Diabetes Clinical Research Program, Benaroya Research Institute, Seattle, Washington 98101
| | - Garth L Warnock
- Department of Surgery, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - Constadina Panagiotopoulos
- Research Institute, BC Children's Hospital, Vancouver, British Columbia V5Z 4H4, Canada
- Department of Medicine, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - C Bruce Verchere
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
- Research Institute, BC Children's Hospital, Vancouver, British Columbia V5Z 4H4, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| |
Collapse
|
21
|
Forst T, Falk A, Andersen G, Fischer A, Weber MM, Voswinkel S, Heise T, Kapitza C, Plum-Mörschel L. Effects on α- and β-cell function of sequentially adding empagliflozin and linagliptin to therapy in people with type 2 diabetes previously receiving metformin: An exploratory mechanistic study. Diabetes Obes Metab 2017; 19:489-495. [PMID: 28009472 DOI: 10.1111/dom.12838] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 11/21/2016] [Accepted: 11/28/2016] [Indexed: 12/20/2022]
Abstract
AIMS To investigate the effect of sequential treatment escalation with empagliflozin and linagliptin on laboratory markers of α- and β-cell function in people with type 2 diabetes mellitus (T2DM) insufficiently controlled on metformin monotherapy. RESEARCH DESIGN AND METHODS A total of 44 people with T2DM received 25 mg empagliflozin for a duration of 1 month in an open-label fashion (treatment period 1 [TP1]). Thereafter, they were randomized to a double-blind add-on therapy with linagliptin 5 mg or placebo (treatment period 2 [TP2]) for 1 additional month. α- and β-cell function was assessed using a standardized liquid meal test and an intravenous (i.v.) glucose challenge. Efficacy measures comprised the areas under the curve for glucose, insulin, proinsulin and glucagon after the liquid meal test and the assessment of fast and late-phase insulin release after an i.v. glucose load with a subsequent hyperglycaemic clamp. RESULTS Empagliflozin reduced fasting and postprandial plasma glucose levels, associated with a significant reduction in postprandial insulin levels and an improvement in the conversion rate of proinsulin (TP1). The addition of linagliptin during TP2 further improved postprandial glucose levels, probably as a result of a marked reduction in postprandial glucagon concentrations (TP2). The insulin response to an i.v. glucose load increased during treatment with empagliflozin (TP1), and further improved after the addition of linagliptin (TP2). CONCLUSION After metformin failure, sequential treatment escalation with empagliflozin and linagliptin is an attractive treatment option because of the additive effects on postprandial glucose control, probably mediated by complementary effects on α- and β-cell function.
Collapse
Affiliation(s)
- Thomas Forst
- Profil, Neuss, Germany
- 1st Medical Department, Endocrinology, Johannes Gutenberg University, Mainz, Germany
| | | | | | | | - Matthias M Weber
- 1st Medical Department, Endocrinology, Johannes Gutenberg University, Mainz, Germany
| | | | | | | | | |
Collapse
|
22
|
Vangipurapu J, Stančáková A, Kuulasmaa T, Kuusisto J, Laakso M. Both fasting and glucose-stimulated proinsulin levels predict hyperglycemia and incident type 2 diabetes: a population-based study of 9,396 Finnish men. PLoS One 2015; 10:e0124028. [PMID: 25853252 PMCID: PMC4390238 DOI: 10.1371/journal.pone.0124028] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 03/09/2015] [Indexed: 01/11/2023] Open
Abstract
Background Hyperproinsulinemia is an indicator of β-cell dysfunction, and fasting proinsulin levels are elevated in patients with hyperglycemia. It is not known whether proinsulin levels after a glucose load are better predictors of hyperglycemia and type 2 diabetes than fasting proinsulin. Methods Participants were 9,396 Finnish men (mean±SD, age 57.3±7.1 years, BMI 27.0±4.0 kg/m2) of the population-based METabolic Syndrome In Men Study who were non-diabetic at the recruitment, and who participated in a 6-year follow-up study. Proinsulin and insulin levels were measured in the fasting state and 30 and 120 min after an oral glucose load. Area under the curve (AUC) and proinsulin to insulin ratios were calculated. Results Fasting proinsulin, proinsulin at 30 min and proinsulin AUC during the first 30 min of an oral glucose tolerance test significantly predicted both the worsening of hyperglycemia and type 2 diabetes after adjustment for confounding factors. Further adjustment for insulin sensitivity (Matsuda index) or insulin secretion (Disposition index) weakened these associations. Insulin sensitivity had a major impact on these associations. Conclusion Our results suggest that proinsulin in the fasting state and after an oral glucose load similarly predict the worsening of hyperglycemia and conversion to type 2 diabetes.
Collapse
Affiliation(s)
- Jagadish Vangipurapu
- Faculty of Health Sciences, Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Alena Stančáková
- Faculty of Health Sciences, Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Teemu Kuulasmaa
- Faculty of Health Sciences, Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Johanna Kuusisto
- Faculty of Health Sciences, Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland; Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Markku Laakso
- Faculty of Health Sciences, Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland; Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
23
|
Li L, Ruau DJ, Patel CJ, Weber SC, Chen R, Tatonetti NP, Dudley JT, Butte AJ. Disease risk factors identified through shared genetic architecture and electronic medical records. Sci Transl Med 2014; 6:234ra57. [PMID: 24786325 DOI: 10.1126/scitranslmed.3007191] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Genome-wide association studies have identified genetic variants for thousands of diseases and traits. We evaluated the relationships between specific risk factors (for example, blood cholesterol level) and diseases on the basis of their shared genetic architecture in a comprehensive human disease-single-nucleotide polymorphism association database (VARIMED), analyzing the findings from 8962 published association studies. Similarity between traits and diseases was statistically evaluated on the basis of their association with shared gene variants. We identified 120 disease-trait pairs that were statistically similar, and of these, we tested and validated five previously unknown disease-trait associations by searching electronic medical records (EMRs) from three independent medical centers for evidence of the trait appearing in patients within 1 year of first diagnosis of the disease. We validated that the mean corpuscular volume is elevated before diagnosis of acute lymphoblastic leukemia; both have associated variants in the gene IKZF1. Platelet count is decreased before diagnosis of alcohol dependence; both are associated with variants in the gene C12orf51. Alkaline phosphatase level is elevated in patients with venous thromboembolism; both share variants in ABO. Similarly, we found that prostate-specific antigen and serum magnesium levels were altered before the diagnosis of lung cancer and gastric cancer, respectively. Disease-trait associations identify traits that could serve as future prognostics, if validated through EMR and subsequent prospective trials.
Collapse
Affiliation(s)
- Li Li
- Division of Systems Medicine, Department of Pediatrics, Stanford University School of Medicine, 1265 Welch Road, Stanford, CA 94305, USA
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Lorenzo C, Hanley AJ, Rewers MJ, Haffner SM. Disproportionately elevated proinsulinemia is observed at modestly elevated glucose levels within the normoglycemic range. Acta Diabetol 2014; 51:617-23. [PMID: 24532116 DOI: 10.1007/s00592-014-0565-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 01/27/2014] [Indexed: 12/31/2022]
Abstract
We aimed to evaluate disproportional proinsulinemia in the pre-diabetic state by analyzing the cross-sectional differences between proinsulin (PI) ratios across the entire range of fasting and 2-h plasma glucose. The study sample was 1,016 participants in the insulin resistance atherosclerosis study, who had no previous diagnosis of diabetes. Insulin sensitivity index (SI) and acute insulin response (AIR) were measured by the frequently sampled intravenous glucose tolerance test. Fasting intact and split PI-to-insulin ratios (PI/I, SPI/I), intact and split PI-to-C-peptide ratios (PI/C-pep, SPI/C-pep), and SI-adjusted AIR were assessed as a function of fasting and 2-h glucose levels. SI-adjusted AIR was decreased (fasting glucose 96-98 mg/dl; 2-h glucose 120-131 mg/dl) and SPI/C-pep increased at modestly elevated fasting glucose and 2-h glucose within the normal glucose tolerance range (fasting glucose 96-98 mg/dl; 2-h glucose 132-142 mg/dl). PI/I was not increased until plasma glucose values were in the diabetic range of fasting glucose (>126 mg/dl) or the impaired glucose tolerance range of 2-h glucose (143-156 mg/dl). SPI/I and PI/C-pep as a function of fasting and 2-h glucose were situated between the curves for SPI/C-pep and PI/I. In conclusion, inappropriate amounts of PI and conversion intermediaries are demonstrated at modestly elevated glucose levels within the normoglycemic range. Ratios that use SPI in the numerator or C-pep in the denominator (and especially SPI/C-pep) are more sensitive to early glycemic excursions than PI/I. Disordered processing of PI may accompany derangements in early insulin secretory response.
Collapse
Affiliation(s)
- Carlos Lorenzo
- Department of Medicine, University of Texas Health Science Center, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA,
| | | | | | | |
Collapse
|
25
|
Insulin regulates carboxypeptidase E by modulating translation initiation scaffolding protein eIF4G1 in pancreatic β cells. Proc Natl Acad Sci U S A 2014; 111:E2319-28. [PMID: 24843127 DOI: 10.1073/pnas.1323066111] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Insulin resistance, hyperinsulinemia, and hyperproinsulinemia occur early in the pathogenesis of type 2 diabetes (T2D). Elevated levels of proinsulin and proinsulin intermediates are markers of β-cell dysfunction and are strongly associated with development of T2D in humans. However, the mechanism(s) underlying β-cell dysfunction leading to hyperproinsulinemia is poorly understood. Here, we show that disruption of insulin receptor (IR) expression in β cells has a direct impact on the expression of the convertase enzyme carboxypeptidase E (CPE) by inhibition of the eukaryotic translation initiation factor 4 gamma 1 translation initiation complex scaffolding protein that is mediated by the key transcription factors pancreatic and duodenal homeobox 1 and sterol regulatory element-binding protein 1, together leading to poor proinsulin processing. Reexpression of IR or restoring CPE expression each independently reverses the phenotype. Our results reveal the identity of key players that establish a previously unknown link between insulin signaling, translation initiation, and proinsulin processing, and provide previously unidentified mechanistic insight into the development of hyperproinsulinemia in insulin-resistant states.
Collapse
|
26
|
Bohl M, Overgaard A, Pietraszek A, Hermansen K. Management of Type 2 diabetes with liraglutide. ACTA ACUST UNITED AC 2014. [DOI: 10.2217/dmt.13.72] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
27
|
Imamura F, Mukamal KJ, Meigs JB, Luchsinger JA, Ix JH, Siscovick DS, Mozaffarian D. Risk factors for type 2 diabetes mellitus preceded by β-cell dysfunction, insulin resistance, or both in older adults: the Cardiovascular Health Study. Am J Epidemiol 2013; 177:1418-29. [PMID: 23707958 DOI: 10.1093/aje/kws440] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Insulin resistance (IR) and pancreatic β-cell dysfunction lead to type 2 diabetes mellitus (DM). We tested whether risk factors would differ for DM that was preceded predominantly by IR, β-cell dysfunction, or both among 4,384 older adults (mean age, 72.7 (standard deviation, 5.6) years) in the Cardiovascular Health Study, which was conducted in North Carolina, California, Maryland, and Pennsylvania (1989-2007). When evaluating established risk factors, we found older age, greater adiposity, higher systolic blood pressure, a lower high-density lipoprotein cholesterol level, a higher triglyceride level, and a lower alcohol intake to be independently associated with greater IR but, conversely, with better β-cell function (P < 0.001). The prospective associations between some risk factors and incident DM varied significantly depending on whether DM was preceded predominantly by IR, β-cell dysfunction, or both. For example, obesity and lower high-density lipoprotein cholesterol levels were positively associated with DM preceded predominantly by IR (hazard ratio (HR) = 5.02, 95% confidence interval (CI): 2.81, 9.00; and HR = 1.97, 95% CI: 1.32, 2.93, respectively), with a significant association with and an insignificant trend toward a lower risk of DM preceded predominantly by β-cell dysfunction (HR = 0.33, 95% CI: 0.14, 0.80; and HR = 0.78, 95% CI: 0.43, 1.39, respectively). In conclusion, among older adults, DM risk factors were differentially associated with DM preceded predominantly by IR or β-cell dysfunction. Biologic and clinical implications of putative subtypes of DM require further investigation.
Collapse
Affiliation(s)
- Fumiaki Imamura
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, USA.
| | | | | | | | | | | | | |
Collapse
|
28
|
Wengert S, Oeztuerk S, Haenle MM, Koenig W, Imhof A, Boehm BO, Wilhelm M, Mao R, Mason RA, Kratzer W. Association of proinsulin and hepatic steatosis in a random, population-based sample. Eur J Endocrinol 2013; 168:195-202. [PMID: 23136404 DOI: 10.1530/eje-12-0605] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Proinsulin may represent a predictive marker for assessing insulin resistance and reduced β-cell function. The objective of this study was to investigate the association between hepatic steatosis, proinsulin and other parameters in a random, population-based sample. DESIGN Cross-sectional study, conducted in south-western Germany. METHODS Upper abdominal ultrasound examinations were performed in 343 subjects (147 females, 196 males; average age 40.0±11.5 years). Proinsulin, the proinsulin-to-insulin ratio and other laboratory parameters were determined, and the BMI, waist-to-hip ratio (WHR) and other anthropometric data were documented. RESULTS HEPATIC STEATOSIS WAS OBSERVED IN 80 SUBJECTS (23.3%: 29.6%, males; 15.0%, females). Multivariate analysis showed an association with hepatic steatosis for male gender (P=0.0212), advancing age (P=0.0241), elevated BMI (P<0.0001), elevated WHR (P=0.0024), alanine aminotransferase (P=0.0046), proinsulin (P=0.0403) and proinsulin-to-insulin ratio (P=0.0116). CONCLUSIONS There is an association between elevated proinsulin concentrations and hepatic steatosis.
Collapse
Affiliation(s)
- Sonja Wengert
- Department of Internal Medicine I, University Hospital Ulm, Albert-Einstein-Allee 23, D-89081 Ulm, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Davalli AM, Perego C, Folli FB. The potential role of glutamate in the current diabetes epidemic. Acta Diabetol 2012; 49:167-83. [PMID: 22218826 DOI: 10.1007/s00592-011-0364-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Accepted: 12/19/2011] [Indexed: 12/27/2022]
Abstract
In the present article, we propose the perspective that abnormal glutamate homeostasis might contribute to diabetes pathogenesis. Previous reports and our recent data indicate that chronically high extracellular glutamate levels exert direct and indirect effects that might participate in the progressive loss of β-cells occurring in both T1D and T2D. In addition, abnormal glutamate homeostasis may impact all the three accelerators of the "accelerator hypothesis" and could partially explain the rising frequency of T1D and T2D.
Collapse
Affiliation(s)
- Alberto M Davalli
- Diabetes and Endocrinology Unit, Department of Internal Medicine, San Raffaele Scientific Institute, 20132, Milan, Italy.
| | | | | |
Collapse
|
30
|
Mercurio V, Carlomagno G, Fazio V, Fazio S. Insulin resistance: Is it time for primary prevention? World J Cardiol 2012; 4:1-7. [PMID: 22279598 PMCID: PMC3262393 DOI: 10.4330/wjc.v4.i1.1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Revised: 12/07/2011] [Accepted: 12/14/2011] [Indexed: 02/06/2023] Open
Abstract
Insulin resistance is a clinical condition characterized by a decrease in sensitivity and responsiveness to the metabolic actions of insulin, so that a given concentration of insulin produces a less-than-expected biological effect. As a result, higher levels of insulin are needed to maintain normal glucose tolerance. Hyperinsulinemia, indeed, is one of the principal characteristics of insulin resistance states. This feature is common in several pathologic conditions, such as type 2 diabetes, obesity, and dyslipidemia, and it is also a prominent component of hypertension, coronary heart disease, and atherosclerosis. The presence of endothelial dysfunction, related to insulin resistance, plays a key role in the development and progression of atherosclerosis in all of these disorders. Insulin resistance represents the earliest detectable abnormality in type 2 diabetes, and is one of the major underlying mechanisms of hypertension and cardiovascular diseases. Its early detection could be of great importance, in order to set a therapeutic attack and to counteract the higher risk of diabetes and cardiovascular diseases.
Collapse
Affiliation(s)
- Valentina Mercurio
- Valentina Mercurio, Guido Carlomagno, Valeria Fazio, Serafino Fazio, Department of Internal Medicine, Cardiovascular and Immunologic Sciences, Federico II University of Naples, 80131 Napoli, Italy
| | | | | | | |
Collapse
|
31
|
Loopstra-Masters RC, Haffner SM, Lorenzo C, Wagenknecht LE, Hanley AJ. Proinsulin-to-C-peptide ratio versus proinsulin-to-insulin ratio in the prediction of incident diabetes: the Insulin Resistance Atherosclerosis Study (IRAS). Diabetologia 2011; 54:3047-54. [PMID: 21959959 DOI: 10.1007/s00125-011-2322-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Accepted: 09/05/2011] [Indexed: 10/17/2022]
Abstract
AIMS Associations of proinsulin-to-insulin ratios with incident type 2 diabetes have been inconsistent. The use of C-peptide as the denominator in the ratio may allow for better prediction because C-peptide concentration is not affected by hepatic insulin clearance. The objective of this paper was to compare fasting intact and split proinsulin-to-insulin ratios (PI/I, SPI/I) with intact and split proinsulin-to-C-peptide ratios (PI/C-pep, SPI/C-pep) in the prediction of type 2 diabetes. METHODS Prospective data on 818 multi-ethnic adults without diabetes at baseline from the Insulin Resistance Atherosclerosis Study (IRAS) were used. Insulin sensitivity (S(I)) and acute insulin response (AIR) were determined from frequently sampled intravenous glucose tolerance tests, and fasting intact and split proinsulin were measured using specific two-site monoclonal antibody-based immunoradiometric assays. Associations of proinsulin ratios with type 2 diabetes were determined using logistic regression and differences in prediction were assessed by comparing areas under the receiver operating characteristic curve (AROCs). RESULTS In logistic regression analyses, PI/C-pep and SPI/C-pep were more strongly associated with incident type 2 diabetes (n = 128) than PI/I and SPI/I, and were significantly better predictors of diabetes in AROC analyses (PI/C-pep = 0.662 vs PI/I = 0.603, p = 0.02; SPI/C-pep = 0.690 vs SPI/I = 0.631, p = 0.01). Both PI/C-pep and SPI/C-pep were associated with type 2 diabetes after adjustment for age, sex, ethnicity, waist circumference, impaired glucose tolerance, lipids and S(I). Both PI/C-pep and SPI/C-pep were significantly associated with incident type 2 diabetes in models that included AIR. CONCLUSIONS Proinsulin-to-C-peptide ratios were stronger predictors of diabetes in comparison with proinsulin-to-insulin ratios. These findings support the use of C-peptide as the denominator for proinsulin ratios, to more accurately reflect the degree of disproportional hyperproinsulinaemia.
Collapse
Affiliation(s)
- R C Loopstra-Masters
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, FitzGerald Building, 150 College St, Toronto, ON, Canada
| | | | | | | | | |
Collapse
|
32
|
Malavolta L, Cabral FR. Peptides: important tools for the treatment of central nervous system disorders. Neuropeptides 2011; 45:309-16. [PMID: 21477861 DOI: 10.1016/j.npep.2011.03.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Revised: 02/11/2011] [Accepted: 03/14/2011] [Indexed: 01/08/2023]
Abstract
This review shows some classical applications of peptides and suggests there is great promise for the treatment of various central nervous system diseases. Actually, peptides are considered the new generation of biologically active tools because they are key regulators in cellular and intercellular physiological responses, which possess enormous potential for the treatment of various diseases. In spite of their clinical potential, native peptides have seen limited use due to their poor bioavailability and low stability in physiological conditions. Moreover, most peptide or protein pharmaceuticals currently in use are delivered by invasive routes such as via subcutaneous injection. Considerable efforts have been made to design new drugs based on peptides and recent developments in technology and science have provided the means and opportunity to produce a stable as well as controlled-release form of peptide and protein drugs to combat poorly controlled diseases and to increase patients' quality of life. A major challenge in this regard, however, is the delivery of peptides over the blood-brain barrier. This review gives an overview of some strategies used to improve both bioavailability and uptake of peptide drugs for delivery into the brain. Indeed, recent findings suggest that the use of peptides by conjugation to a polymer such as nanoparticles can offer tremendous hope in the treatment of brain disorders. The polymer conjugation improves pharmacokinetics by increasing the molecular mass of proteins and peptides and shielding them from proteolytic enzymes. These new strategies will create new opportunities for the future development of neurotherapeutic drugs. In the present review we have focused our attention on the peptide controlled delivery, summarizing literature reports on the use of peptides and nanotechnology for the treatment and diagnosis of brain disorders.
Collapse
Affiliation(s)
- Luciana Malavolta
- Brain Institute-Instituto Israelita de Ensino e Pesquisa Albert Einstein, Morumbi-São Paulo, SP, Brazil.
| | | |
Collapse
|
33
|
Dual and opposing roles of the unfolded protein response regulated by IRE1alpha and XBP1 in proinsulin processing and insulin secretion. Proc Natl Acad Sci U S A 2011; 108:8885-90. [PMID: 21555585 DOI: 10.1073/pnas.1105564108] [Citation(s) in RCA: 215] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
As a key regulator of the unfolded protein response, the transcription factor XBP1 activates genes in protein secretory pathways and is required for the development of certain secretory cells. To elucidate the function of XBP1 in pancreatic β-cells, we generated β-cell-specific XBP1 mutant mice. Xbp1(f/f);RIP-cre mice displayed modest hyperglycemia and glucose intolerance resulting from decreased insulin secretion from β-cells. Ablation of XBP1 markedly decreased the number of insulin granules in β-cells, impaired proinsulin processing, increased the serum proinsulin:insulin ratio, blunted glucose-stimulated insulin secretion, and inhibited cell proliferation. Notably, XBP1 deficiency not only compromised the endoplasmic reticulum stress response in β-cells but also caused constitutive hyperactivation of its upstream activator, IRE1α, which could degrade a subset of mRNAs encoding proinsulin-processing enzymes. Hence, the combined effects of XBP1 deficiency on the canonical unfolded protein response and its negative feedback activation of IRE1α caused β-cell dysfunction in XBP1 mutant mice. These results demonstrate that IRE1α has dual and opposing roles in β-cells, and that a precisely regulated feedback circuit involving IRE1α and its product XBP1s is required to achieve optimal insulin secretion and glucose control.
Collapse
|
34
|
Di Cairano ES, Davalli AM, Perego L, Sala S, Sacchi VF, La Rosa S, Finzi G, Placidi C, Capella C, Conti P, Centonze VE, Casiraghi F, Bertuzzi F, Folli F, Perego C. The glial glutamate transporter 1 (GLT1) is expressed by pancreatic beta-cells and prevents glutamate-induced beta-cell death. J Biol Chem 2011; 286:14007-18. [PMID: 21335552 DOI: 10.1074/jbc.m110.183517] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Glutamate is the major excitatory neurotransmitter of the central nervous system (CNS) and may induce cytotoxicity through persistent activation of glutamate receptors and oxidative stress. Its extracellular concentration is maintained at physiological concentrations by high affinity glutamate transporters of the solute carrier 1 family (SLC1). Glutamate is also present in islet of Langerhans where it is secreted by the α-cells and acts as a signaling molecule to modulate hormone secretion. Whether glutamate plays a role in islet cell viability is presently unknown. We demonstrate that chronic exposure to glutamate exerts a cytotoxic effect in clonal β-cell lines and human islet β-cells but not in α-cells. In human islets, glutamate-induced β-cell cytotoxicity was associated with increased oxidative stress and led to apoptosis and autophagy. We also provide evidence that the key regulator of extracellular islet glutamate concentration is the glial glutamate transporter 1 (GLT1). GLT1 localizes to the plasma membrane of β-cells, modulates hormone secretion, and prevents glutamate-induced cytotoxicity as shown by the fact that its down-regulation induced β-cell death, whereas GLT1 up-regulation promoted β-cell survival. In conclusion, the present study identifies GLT1 as a new player in glutamate homeostasis and signaling in the islet of Langerhans and demonstrates that β-cells critically depend on its activity to control extracellular glutamate levels and cellular integrity.
Collapse
Affiliation(s)
- Eliana S Di Cairano
- Department of Molecular Science Applied to Biosystems, Università degli Studi di Milano, 20134 Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Byrd-Williams CE, Strother ML, Kelly LA, Huang TTK. Dietary fiber and associations with adiposity and fasting insulin among college students with plausible dietary reports. Nutrition 2009; 25:896-904. [PMID: 19403267 PMCID: PMC2831808 DOI: 10.1016/j.nut.2009.02.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2008] [Revised: 02/04/2009] [Accepted: 02/24/2009] [Indexed: 10/20/2022]
Abstract
OBJECTIVE We examined dietary fiber intake, food sources of dietary fiber, and relation of dietary fiber to body composition and metabolic parameters in college students with plausible dietary reports. METHODS Students (18-24 y of age) provided data on anthropometry, fasting blood chemistries, and body composition (bioelectric impedance). Diet and physical activity were assessed with the Diet History Questionnaire and the International Physical Activity Questionnaire. Plausible dietary reporters were identified (+/-1 SD cutoffs for reported energy intake as a percentage of predicted energy requirement). Multiple regression analyses were conducted with the total (n = 298) and plausible (n = 123) samples, adjusting for age, race, sex, smoking status, physical activity, energy intake, and fat-free mass (where applicable). RESULTS Food sources of dietary fiber were similar in men and women. In the plausible sample compared with the total sample, dietary fiber was more strongly associated with fat mass (beta = -0.24, P < 0.001), percentage of body fat (beta = -0.23, P < 0.001), body mass index (beta = -0.11, P < 0.01), waist circumference (beta = -0.67, P < 0.05), and fasting insulin (beta = -0.15, P < 0.001). When the effect of sex was investigated, dietary fiber was inversely related to fasting insulin and fat mass in men and women and inversely related to percentage of body fat, body mass index, and waist circumference in men only (P < 0.05). CONCLUSION Inclusion of implausible dietary reports may result in spurious or weakened diet-health associations. Dietary fiber is negatively associated with fasting insulin levels in men and women and consistently associated with adiposity measurements in men.
Collapse
Affiliation(s)
- Courtney E Byrd-Williams
- Department of Preventive Medicine, University of Southern California, Los Angeles, California, USA
| | | | | | | |
Collapse
|
36
|
Petursson P, Herlitz J, Caidahl K, From-Attebring M, Sjöland H, Gudbjörnsdottir S, Hartford M. Association between glycometabolic status in the acute phase and 2½ years after an acute coronary syndrome. SCAND CARDIOVASC J 2009; 40:145-51. [PMID: 16798661 DOI: 10.1080/14017430600797626] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVES To evaluate the association between glycometabolic status in the acute phase and 21/2 years later in patients with acute coronary syndrome (ACS). METHODS Non-diabetic patients (n = 762) presenting with ACS were prospectively followed up for 21/2 years. Patients were stratified by admission plasma glucose (<6.1 mmol/l, 6.1 - 6.9 mmol/l and >or=7.0 mmol/l) and HbA1c (<or=4.5%, 4.6 - 5.4% and >or=5.5%). The predictive value of glucose levels >or= 7.0 mmol/l and HbA1c >or= 5.5% for glycometabolic disturbance (i.e. diabetes or impaired fasting glycaemia (IFG)) was analysed. RESULTS Of 762 patients, 13% had a diagnosis of diabetes and 16% had IFG at follow-up. The prevalence of glycometabolic disturbance at follow-up increased with increasing plasma glucose at admission, from 19% in patients with < 6.1 mmol/l to 42% in patients with >or= 7.0 mmol/l. Sixty-one percent of patients with HbA1c >or= 5.5% had glycometabolic disturbance after 21/2 years compared to only 25% of those with HbA1c < 5.5%. CONCLUSION Non-diabetic patients with ACS and hyperglycaemia are at high risk for developing glycometabolic disturbance. HbA1c may be an even stronger predictor of glycometabolic disturbance than plasma glucose.
Collapse
Affiliation(s)
- P Petursson
- Department of Cardiology, Sahlgrenska University Hospital, Göteborg, Sweden.
| | | | | | | | | | | | | |
Collapse
|
37
|
Ferrannini E, Muscelli E, Natali A, Gabriel R, Mitrakou A, Flyvbjerg A, Golay A, Hojlund K. Association of fasting glucagon and proinsulin concentrations with insulin resistance. Diabetologia 2007; 50:2342-7. [PMID: 17846745 DOI: 10.1007/s00125-007-0806-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2007] [Accepted: 07/24/2007] [Indexed: 01/29/2023]
Abstract
AIMS/HYPOTHESIS Hyperproinsulinaemia and relative hyperglucagonaemia are features of type 2 diabetes. We hypothesised that raised fasting glucagon and proinsulin concentrations may be associated with insulin resistance (IR) in non-diabetic individuals. METHODS We measured IR [by a euglycaemic-hyperinsulinaemic (240 pmol min(-1) m(-2)) clamp technique] in 1,296 non-diabetic (on a 75 g OGTT) individuals [716 women and 579 men, mean age 44 years, BMI 26 kg/m(2) (range 18-44 kg/m(2))] recruited at 19 centres in 14 European countries. IR was related to fasting proinsulin or pancreatic glucagon concentrations in univariate and multivariate analyses. Given its known relationship to IR, serum adiponectin was used as a positive control. RESULTS In either sex, both glucagon and proinsulin were directly related to IR, while adiponectin was negatively associated with it (all p < 0.0001). In multivariate models, controlling for known determinants of insulin sensitivity (i.e. sex, age, BMI and glucose tolerance) as well as factors potentially affecting glucagon and proinsulin (i.e. fasting plasma glucose and C-peptide concentrations), glucagon and proinsulin were still positively associated, and adiponectin was negatively associated, with IR. Finally, when these associations were tested as the probability that individuals in the top IR quartile would have hormone levels in the top quartile of their distribution independently of covariates, the odds ratio was approximately 2 for both glucagon (p = 0.05) and proinsulin (p = 0.02) and 0.36 for adiponectin (p < 0.0001). CONCLUSIONS/INTERPRETATION Whole-body IR is independently associated with raised fasting plasma glucagon and proinsulin concentrations, possibly as a result of IR at the level of alpha cells and beta cells in pancreatic islets.
Collapse
Affiliation(s)
- E Ferrannini
- Department of Internal Medicine and CNR Institute of Clinical Physiology, University of Pisa, Pisa, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Pivatto I, Bustos P, Amigo H, Acosta AM, Arteaga A. Association between proinsulin, insulin, proinsulin/insulin ratio, and insulin resistance status with the metabolic syndrome. ACTA ACUST UNITED AC 2007; 51:1128-33. [DOI: 10.1590/s0004-27302007000700016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2006] [Accepted: 04/12/2007] [Indexed: 11/21/2022]
Abstract
The Metabolic Syndrome (MS) constitutes an independent risk factor of cardiovascular disease. There is evidence that proinsulin blood levels and the proinsulin/insulin ratio are associated to the MS. The purpose of this study was to compare proinsulin and insulin, insulin resistance index, and the proinsulin/insulin ratio as predictors of MS. This is a cross-sectional study involving 440 men and 556 women with a mean age of 24 years. Diagnosis of MS was made according to the National Cholesterol Education Program Adult Treatment Panel III. Blood levels of insulin and proinsulin were measured, and the insulin resistance status was estimated using the homeostatic model assessment (HOMA-IR). The prevalence of MS was 10.1%. HOMA-IR was the best MS risk factor for both women and men (OR = 2.04; 95% CI: 1.68-2.48 and 1.09; 95% CI: 1.05-1.13, respectively). HOMA-IR presented the best positive predictive value for MS: 22% and 36% for men and women, respectively, and was the best MS indicator. The proinsulin/insulin ratio did not show significant association with MS. HOMA-IR, proinsulin, and insulin presented good negative predictive values for both genders that could be used to identify an at-risk population.
Collapse
Affiliation(s)
| | - Patricia Bustos
- Pontifical Catholic University of Chile; University of Chile, Chile
| | - Hugo Amigo
- Pontifical Catholic University of Chile; University of Chile, Chile
| | | | | |
Collapse
|
39
|
Abstract
The insulin resistance syndrome, also referred to as the metabolic syndrome or syndrome X, is associated with a primary cellular defect in insulin action (insulin resistance) and a compensatory increase in insulin secretion. The combination of insulin resistance and subsequent hyperinsulinaemia causes a number of metabolic and cardiovascular changes that result in a syndrome typically characterised by type 2 diabetes, obesity, dyslipidaemia, coronary artery disease and hypertension. Moreover, disturbances in sleep (sleep apnoea) and ovarian dysfunction are also characterised by insulin resistance. The pathophysiological basis for these disturbances reflects the impact of variable genetic and environmental influences. At a molecular level, insulin resistance involves defects of insulin signalling such as reduced insulin receptor tyrosine kinase activity and reduced post-receptor phosphorylation steps that impinge on metabolic and vascular effects of insulin.
Collapse
Affiliation(s)
- Sangeeta R Kashyap
- Department of Endocrinology, Diabetes and Metabolism, Cleveland Clinic Foundation, Cleveland, OH 44195, USA.
| | | |
Collapse
|
40
|
Abstract
BACKGROUND Type 2 diabetes results from increasing insulin resistance coupled with progressive loss of beta-cell function. Further deterioration of beta-cell function is associated with progression of diabetes and the potential development of microvascular and macrovascular complications. SCOPE This review examines current knowledge of beta-cell function and uses this information to assess how the results of ongoing trials could increase our understanding of disease progression and potential interventions. Data were derived from a Medline search using the search terms 'beta-cell dysfunction', 'IGT', 'thiazolidinediones', 'metformin', and 'sulfonylurea'. RESULTS The mechanisms that underlie beta-cell dysfunction are complex and most likely involve the interplay of a range of factors that reduce both beta-cell mass and secretory function. These include detrimental effects associated with hyperglycemia itself, elevated free fatty acids, and inflammatory responses linked to adipocyte-derived cytokines, with apoptosis a key underlying mechanism. Early intervention with treatments that address these defects, and preserve beta-cell function while improving insulin sensitivity, may delay disease progression in patients with type 2 diabetes and also prevent the development of diabetes in 'at-risk' individuals. Two of the studies examining the effects of interventions on development and progression of diabetes that are due to report shortly are ADOPT (A Diabetes Outcome Progression Trial), comparing a thiazolidinedione, metformin and a sulfonylurea in patients with type 2 diabetes not previously treated with oral hypoglycemic agents, and DREAM (Diabetes REduction Assessment with ramipril and rosiglitazone Medication), assessing the effects of a thiazolidinedione and an angiotensin-converting enzyme inhibitor in subjects with impaired glucose tolerance and/or impaired fasting glucose. CONCLUSIONS Although we do not have a full understanding of the mechanisms driving progression of type 2 diabetes, there is growing evidence that we may be able to modulate them and thereby improve patient outcomes.
Collapse
Affiliation(s)
- Lawrence A Leiter
- Department of Medicine, University of Toronto, St. Michael's Hospital, Toronto, ON, Canada.
| |
Collapse
|
41
|
Pradhan AD, Manson JE, Hendrix SL, Johnson KC, Wagenknecht LE, Haan MN, Weidner G, Lacroix AZ, Cook NR. Cross-sectional correlates of fasting hyperinsulinaemia in post-menopausal women of different ethnic origin. Diabet Med 2006; 23:77-85. [PMID: 16409570 DOI: 10.1111/j.1464-5491.2006.01788.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
AIMS In a large ethnically diverse nationwide sample of post-menopausal women we explored the relationship between fasting insulin levels, ethnicity, and a wide range of anthropometric, socio-economic, and lifestyle factors. METHODS Subjects were post-menopausal women aged 50-79 years without diagnosed diabetes mellitus comprising a subsample (n = 3500) of the Women's Health Initiative (WHI) Clinical Trial and Observational Study. In a cross-sectional survey at baseline, we analysed the association between ethnicity and fasting insulin using analysis of covariance procedures and identified independent correlates of hyperinsulinaemia, defined by the 75th percentile cut point for each ethnic group. RESULTS Fasting insulin levels were higher among African-American and Hispanic women than among non-Hispanic White or Asian women. These differences persisted after adjustment for age, educational attainment, total and central body obesity, adult weight change, family history of diabetes, smoking status, alcohol consumption, use of menopausal hormone therapy and physical activity. Higher levels of body mass index, waist-hip ratio, adult weight gain, and lower levels of total and moderate or strenuous recreational activity were independent correlates of fasting hyperinsulinaemia. Habitual walking was also inversely associated with fasting insulin. CONCLUSIONS In this cross-sectional analysis, fasting insulin levels were higher among African-American and Hispanic post-menopausal women as compared with non-Hispanic White and Asian women. In addition, obesity, adult weight gain, and low levels of moderate or strenuous physical activity were independently associated with hyperinsulinaemia.
Collapse
Affiliation(s)
- A D Pradhan
- Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Bak MI, Grochowiecki T, Gałazka Z, Nazarewski S, Jakimowicz T, Pietrasik K, Wojtaszek M, Durlik M, Karnafel W, Szmidt J. Proinsulinemia in Simultaneous Pancreas and Kidney Transplant Recipients. Transplant Proc 2006; 38:280-1. [PMID: 16504725 DOI: 10.1016/j.transproceed.2005.12.037] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
It has been shown that lipid profiles do not differ between pancreas recipients with systemic and portal venous anastomosis. However, it is unclear whether venous drainage from the transplanted pancreas has an impact on recipient atherogenesis and if other factors should be considered. Increased concentration of proinsulin correlates with tachycardia and other risk factors for ischemic heart disease. The aim of this study was to compare proinsulin levels in different types of pancreatic graft venous drainage. Twenty-four simultaneous pancreas and kidney transplantation (SPK) recipients with systemic venous drainage (group S, n = 12) and portal venous drainage (group P, n = 12) under identical immunosuppressive treatment were prospectively observed during 24 months. Following transplantation, only recipients with normoglycemia, normal HbA1c, and normal serum creatine were evaluated. Proinsulin was assessed in fasting state; after glucagon stimulation (Delta-proinsulin), and during oral 75-g glucose tolerance test twice: between 3 and 6 months and 12 to 24 months posttransplantation. All SPK patients had higher proinsulin concentration in fasting state compared with age-matched healthy controls. After stimulation, proinsulin level did not significantly differ between groups; the type of the pancreas venous anastomosis did not change the release of proinsulin and should not have impact on cardiovascular risk factors.
Collapse
Affiliation(s)
- M I Bak
- Department of Gastroenterology and Metabolic Diseases, Warsaw Medical University, ul. Banacha 1a, 02-097 Warsaw, Poland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Dessein PH, Joffe BI, Stanwix AE. Editorial: should we evaluate insulin sensitivity in rheumatoid arthritis? Semin Arthritis Rheum 2005; 35:5-7. [PMID: 16084218 DOI: 10.1016/j.semarthrit.2005.03.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
44
|
Retnakaran R, Hanley AJG, Sermer M, Zinman B. The impact of insulin resistance on proinsulin secretion in pregnancy: hyperproinsulinemia is not a feature of gestational diabetes. Diabetes Care 2005; 28:2710-5. [PMID: 16249544 DOI: 10.2337/diacare.28.11.2710] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Excessive secretion of the insulin precursor proinsulin, as manifested by an increased serum proinsulin-to-insulin ratio, has been associated with beta-cell dysfunction. In women with gestational diabetes mellitus (GDM), previous studies of the proinsulin-to-insulin ratio have yielded conflicting results, despite the presence of beta-cell dysfunction. The interpretation of the proinsulin-to-insulin ratio, however, may be confounded by the variable effects of hepatic insulin extraction. Thus, we sought to determine whether GDM is characterized by relative hyperproinsulinemia as measured by the proinsulin-to-C-peptide ratio, an alternate measure of proinsulin secretion that is not affected by hepatic insulin extraction. RESEARCH DESIGN AND METHODS Serum proinsulin, C-peptide, and insulin were measured in a cross-sectional study of 180 women undergoing oral glucose tolerance tests (OGTTs) in the late second or early third trimester. Based on the OGTT, participants were stratified into three groups: 1) normal glucose tolerance (NGT; n = 93), 2) impaired glucose tolerance (IGT; n = 39), and 3) GDM (n = 48). Insulin sensitivity (IS) was measured using the IS(OGTT) index of Matsuda and DeFronzo, which has been previously validated in pregnant women. RESULTS There were no significant differences in mean fasting proinsulin-to-C-peptide ratio between the three glucose tolerance groups (NGT, 0.024; IGT, 0.022; GDM, 0.019; P = 0.4). Furthermore, adjustment for age, weeks' gestation, prepregnancy BMI, ethnicity, previous GDM, and family history of diabetes did not reveal any association between the proinsulin-to-C-peptide ratio and glucose tolerance status. Using Spearman univariate correlation analysis, fasting proinsulin-to-C-peptide ratio was significantly correlated with IS(OGTT) (r = 0.29, P < 0.0001) and inversely related to the homeostasis model assessment of insulin resistance (r = -0.36, P < 0.0001) and prepregnancy BMI (r = -0.23, P < 0.005). On multiple linear regression analysis, IS(OGTT) emerged as the strongest independent correlate of the dependent variable proinsulin-to-C-peptide ratio. Furthermore, after adjustment for potential covariates, a stepwise decrease in proinsulin-to-C-peptide ratio was observed per decreasing tertile of IS(OGTT) (trend P = 0.0019), consistent with enhanced efficiency of proinsulin processing (i.e., reduced proinsulin-to-C-peptide ratio) as insulin resistance increases. CONCLUSIONS GDM is not independently associated with hyperproinsulinemia as measured by the proinsulin-to-C-peptide ratio. Instead, in pregnant women, increased insulin resistance is associated with decreased proinsulin-to-C-peptide ratio, independently of glucose tolerance status. These data suggest that relative proinsulin secretion in late pregnancy is primarily related to insulin resistance and does not necessarily reflect beta-cell function.
Collapse
Affiliation(s)
- Ravi Retnakaran
- Division of Endocrinology, University of Toronto, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
45
|
Schulze MB, Solomon CG, Rifai N, Cohen RM, Sparrow J, Hu FB, Manson JE. Hyperproinsulinaemia and risk of Type 2 diabetes mellitus in women. Diabet Med 2005; 22:1178-84. [PMID: 16108846 DOI: 10.1111/j.1464-5491.2005.01585.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
AIMS Our objective was to examine prospectively the associations between fasting plasma proinsulin and the proinsulin/insulin ratio and the incidence of Type 2 diabetes in women. SUBJECTS AND METHODS We designed a nested case-control study within the Nurses' Health Study, a cohort of 121,700 US women aged 30-55 years at study inception in 1976. Fasting plasma proinsulin, specific insulin and C-peptide levels were determined in 183 women with a new diagnosis of diabetes made after blood sampling between 1989 and 1990, and 369 control subjects without diabetes. RESULTS After adjustment for age, body mass index, family history of diabetes and other potential confounders, including HbA1c, the odds ratios for diabetes associated with increasing quartiles of proinsulin were 1.00, 0.85, 2.49 and 5.73 (P for trend: < 0.001). Proinsulin remained significantly associated with diabetes risk after adjusting for C-peptide and specific insulin (multivariate odds ratios for quartiles: 1.00, 0.78, 1.94, 3.69; P for trend = 0.001). In addition, the proinsulin/insulin ratio was significantly associated with diabetes risk, controlling in multivariate analysis for C-peptide (odds ratios for extreme quartiles: 2.48; 95% CI: 1.14-5.41; P for trend = 0.005). CONCLUSIONS These data suggest that proinsulin and the proinsulin/insulin ratio are strong independent predictors of diabetes risk, after adjustment for obesity and other potential confounders.
Collapse
Affiliation(s)
- M B Schulze
- Department of Nutrition, Harvard School of Public Health, Boston, MA, USA.
| | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
Type 2 diabetes is caused by progressively increasing insulin resistance coupled with deteriorating beta-cell function, and there is a growing body of evidence to suggest that both of these defects precede hyperglycaemia by many years. Several studies have demonstrated the importance of maintaining beta-cell function in patients with Type 2 diabetes. This review explores parameters used to indicate beta-cell dysfunction, in Type 2 diabetes and in individuals with a predisposition to the disease. A genetic element undoubtedly underlies beta-cell dysfunction; however, a number of modifiable components are also associated with beta-cell deterioration, such as chronic hyperglycaemia and elevated free fatty acids. There is also evidence for a link between pro-inflammatory cytokines and impairment of insulin-signalling pathways in the beta-cell, and the potential role of islet amyloid deposition in beta-cell deterioration continues to be a subject for debate. The thiazolidinediones are a class of agents that have demonstrated clinical improvements in indices of beta-cell dysfunction and have the potential to improve beta-cell function. Data are accumulating to show that this therapeutic group offers a number of advantages over traditionally employed oral agents, and these data demonstrate the growing importance of thiazolidinediones in Type 2 diabetes management.
Collapse
Affiliation(s)
- L A Leiter
- Department of Medicine, University of Toronto, St. Michael's Hospital, Toronto, Ontario, Canada.
| |
Collapse
|
47
|
Giles TD, Sander GE. Diabetes mellitus and heart failure: basic mechanisms, clinical features, and therapeutic considerations. Cardiol Clin 2005; 22:553-68. [PMID: 15501623 DOI: 10.1016/j.ccl.2004.07.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Diabetic cardiomyopathy encompasses the spectrum from subclinical disease to the full-blown syndrome of congestive heart failure. The prevalence of type 2 diabetes mellitus is increasing at an alarming rate in the western world. and with it, the frequency of diabetes-related heart failure. There is at least early suggestion that target-driven, long-term, intensified intervention that is aimed at multiple risk factors in patients who have type 2 diabetes and microalbuminuria may reduce the risk of macrovascular (cardiovascular) and micro-vascular complications by approximately 50%. Thus, it is imperative that patients, particularly those who are at risk for the cardiovascular dysmetabolic syndrome, be screened aggressively for the presence of glucose intolerance and diabetes. When detected, all metabolic and cardio-vascular parameters should be evaluated and treated aggressively to reach currently recommended clinical targets. Such action will result in great benefit for patients by reducing morbidity and mortality and improving quality of life and will reduce the financial burden that is associated with this epidemic disease.
Collapse
Affiliation(s)
- Thomas D Giles
- Section of Cardiology, Louisiana State University Health Sciences Center, 1542 Tulane Avenue, New Orleans, LA 70112, USA.
| | | |
Collapse
|
48
|
Del Prato S, Marchetti P. Targeting insulin resistance and beta-cell dysfunction: the role of thiazolidinediones. Diabetes Technol Ther 2004; 6:719-31. [PMID: 15628822 DOI: 10.1089/dia.2004.6.719] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Insulin resistance and beta-cell dysfunction are fundamental defects that contribute to the development of type 2 diabetes, and as such are targets for primary prevention of disease progression. The two parameters are linked by several factors, including glucotoxicity and lipotoxicity, and recent research has enlightened understanding of the molecular mechanisms underlying the development and progression of the disease. Historically, type 2 diabetes has been managed by controlling hyperglycemia, using agents that increase insulin levels or reduce hepatic glucose production, as exemplified by the United Kingdom Prospective Diabetes Study. The thiazolidinediones control hyperglycemia by targeting the fundamental defects of the disease, and have shown well-documented improvements in insulin sensitivity and beta-cell function, both in monotherapy and in combination with other oral antidiabetic agents. TRoglitazone In the Prevention Of Diabetes (TRIPOD) has demonstrated the potential for thiazolidinediones to delay progression to type 2 diabetes. Prospective studies such as Diabetes REduction Approaches with ramipril and rosiglitazone Medications (DREAM) are currently evaluating the long-term effects of thiazolidinediones on metabolic status and disease progression.
Collapse
Affiliation(s)
- S Del Prato
- Section of Diabetes and Metabolic Diseases, Department of Endocrinology and Metabolism, Ospedale Cisanello, Pisa, Italy.
| | | |
Collapse
|
49
|
McLaughlin TL, Reaven GM. Beyond type 2 diabetes: the need for a clinically useful way to identify insulin resistance. Am J Med 2003; 114:501-2. [PMID: 12727584 DOI: 10.1016/s0002-9343(03)00122-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|