1
|
Hurtado-Alvarado G, Soto-Tinoco E, Santacruz-Martínez E, Prager-Khoutorsky M, Escobar C, Buijs RM. Suprachiasmatic nucleus promotes hyperglycemia induced by sleep delay. Curr Biol 2023; 33:4343-4352.e4. [PMID: 37725978 DOI: 10.1016/j.cub.2023.08.071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/05/2023] [Accepted: 08/23/2023] [Indexed: 09/21/2023]
Abstract
Short sleep is linked to disturbances in glucose metabolism and may induce a prediabetic condition. The biological clock in the suprachiasmatic nucleus (SCN) regulates the glucose rhythm in the circulation and the sleep-wake cycle. SCN vasopressin neurons (SCNVP) control daily glycemia by regulating the entrance of glucose into the arcuate nucleus (ARC). Thus, we hypothesized that sleep delay may influence SCN neuronal activity. We, therefore, investigated the role of SCNVP when sleep is disrupted by forced locomotor activity. After 2 h of sleep delay, rats exhibited decreased SCNVP neuronal activity, a decrease in the glucose transporter GLUT1 expression in tanycytes lining the third ventricle, lowered glucose entrance into the ARC, and developed hyperglycemia. The association between reduced SCNVP neuronal activity and hyperglycemia in sleep-delayed rats was evidenced by injecting intracerebroventricular vasopressin; this increased GLUT1 immunoreactivity in tanycytes, thus promoting normoglycemia. Following sleep recovery, glucose levels decreased, whereas SCNVP neuronal activity increased. These results imply that sleep-delay-induced changes in SCNVP activity lead to glycemic impairment, inferring that disruption of biological clock function might represent a critical step in developing type 2 diabetes.
Collapse
Affiliation(s)
- Gabriela Hurtado-Alvarado
- Department of Cellular Biology and Physiology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mario de la Cueva Circuit, 04510 Mexico City, Mexico
| | - Eva Soto-Tinoco
- Department of Cellular Biology and Physiology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mario de la Cueva Circuit, 04510 Mexico City, Mexico
| | - Esteban Santacruz-Martínez
- Department of Cellular Biology and Physiology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mario de la Cueva Circuit, 04510 Mexico City, Mexico
| | - Masha Prager-Khoutorsky
- Department of Physiology, McIntyre Medical Sciences Building, McGill University, 3655 Promenade Sir-William-Osler, Montréal, QC H3G 1Y6, Canada
| | - Carolina Escobar
- Departamento de Anatomía, Facultad de Medicina, Universidad Nacional Autónoma de México, 04510 Mexico City, Mexico
| | - Ruud M Buijs
- Department of Cellular Biology and Physiology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mario de la Cueva Circuit, 04510 Mexico City, Mexico.
| |
Collapse
|
2
|
Ahn J, Baik JW, Kim D, Choi K, Lee S, Park SM, Kim JY, Nam SH, Kim C. In vivo photoacoustic monitoring of vasoconstriction induced by acute hyperglycemia. PHOTOACOUSTICS 2023; 30:100485. [PMID: 37082618 PMCID: PMC10112177 DOI: 10.1016/j.pacs.2023.100485] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 03/19/2023] [Accepted: 03/29/2023] [Indexed: 05/03/2023]
Abstract
Postprandial hyperglycemia, blood glucose spikes, induces endothelial dysfunction, increasing cardiovascular risks. Endothelial dysfunction leads to vasoconstriction, and observation of this phenomenon is important for understanding acute hyperglycemia. However, high-resolution imaging of microvessels during acute hyperglycemia has not been fully developed. Here, we demonstrate that photoacoustic microscopy can noninvasively monitor morphological changes in blood vessels of live animals' extremities when blood glucose rises rapidly. As blood glucose level rose from 100 to 400 mg/dL following intraperitoneal glucose injection, heart/breath rate, and body temperature remained constant, but arterioles constricted by approximately -5.7 ± 1.1% within 20 min, and gradually recovered for another 40 min. In contrast, venular diameters remained within about 0.6 ± 1.5% during arteriolar constriction. Our results experimentally and statistically demonstrate that acute hyperglycemia produces transitory vasoconstriction in arterioles, with an opposite trend of change in blood glucose. These findings could help understanding vascular glucose homeostasis and the relationship between diabetes and cardiovascular diseases.
Collapse
Affiliation(s)
- Joongho Ahn
- Departments of Electrical Engineering, Convergence IT Engineering, Mechanical Engineering, and Medical Science and Engineering, and Medical Device Innovation Center, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Jin Woo Baik
- Departments of Electrical Engineering, Convergence IT Engineering, Mechanical Engineering, and Medical Science and Engineering, and Medical Device Innovation Center, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Donggyu Kim
- Departments of Electrical Engineering, Convergence IT Engineering, Mechanical Engineering, and Medical Science and Engineering, and Medical Device Innovation Center, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Karam Choi
- Samsung Advanced Institute of Technology, Samsung Electronics Co. Ltd., Suwon 16678, Republic of Korea
| | - Seunghyun Lee
- Departments of Electrical Engineering, Convergence IT Engineering, Mechanical Engineering, and Medical Science and Engineering, and Medical Device Innovation Center, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Sung-Min Park
- Departments of Electrical Engineering, Convergence IT Engineering, Mechanical Engineering, and Medical Science and Engineering, and Medical Device Innovation Center, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Jin Young Kim
- Departments of Electrical Engineering, Convergence IT Engineering, Mechanical Engineering, and Medical Science and Engineering, and Medical Device Innovation Center, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Sung Hyun Nam
- Samsung Advanced Institute of Technology, Samsung Electronics Co. Ltd., Suwon 16678, Republic of Korea
- Corresponding authors.
| | - Chulhong Kim
- Departments of Electrical Engineering, Convergence IT Engineering, Mechanical Engineering, and Medical Science and Engineering, and Medical Device Innovation Center, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
- Corresponding authors.
| |
Collapse
|
3
|
Sequeira IR, Yip W, Lu LW, Jiang Y, Murphy R, Plank LD, Cooper GJS, Peters CN, Aribsala BS, Hollingsworth KG, Poppitt SD. Exploring the relationship between pancreatic fat and insulin secretion in overweight or obese women without type 2 diabetes mellitus: A preliminary investigation of the TOFI_Asia cohort. PLoS One 2022; 17:e0279085. [PMID: 36584200 PMCID: PMC9803309 DOI: 10.1371/journal.pone.0279085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 10/02/2022] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE While there is an emerging role of pancreatic fat in the aetiology of type 2 diabetes mellitus (T2DM), its impact on the associated decrease in insulin secretion remains controversial. We aimed to determine whether pancreatic fat negatively affects β-cell function and insulin secretion in women with overweight or obesity but without T2DM. METHODS 20 women, with normo- or dysglycaemia based on fasting plasma glucose levels, and low (< 4.5%) vs high (≥ 4.5%) magnetic resonance (MR) quantified pancreatic fat, completed a 1-hr intravenous glucose tolerance test (ivGTT) which included two consecutive 30-min square-wave steps of hyperglycaemia generated by using 25% dextrose. Plasma glucose, insulin and C-peptide were measured, and insulin secretion rate (ISR) calculated using regularisation deconvolution method from C-peptide kinetics. Repeated measures linear mixed models, adjusted for ethnicity and baseline analyte concentrations, were used to compare changes during the ivGTT between high and low percentage pancreatic fat (PPF) groups. RESULTS No ethnic differences in anthropomorphic variables, body composition, visceral adipose tissue (MR-VAT) or PPF were measured and hence data were combined. Nine women (47%) were identified as having high PPF values. PPF was significantly associated with baseline C-peptide (p = 0.04) and ISR (p = 0.04) in all. During the 1-hr ivGTT, plasma glucose (p<0.0001), insulin (p<0.0001) and ISR (p = 0.02) increased significantly from baseline in both high and low PPF groups but did not differ between the two groups at any given time during the test (PPF x time, p > 0.05). Notably, the incremental areas under the curves for both first and second phase ISR were 0.04 units lower in the high than low PPF groups, but this was not significant (p > 0.05). CONCLUSION In women with overweight or obesity but without T2DM, PPF did not modify β-cell function as determined by ivGTT-assessed ISR. However, the salient feature in biphasic insulin secretion in those with ≥4.5% PPF may be of clinical importance, particularly in early stages of dysglycaemia may warrant further investigation.
Collapse
Affiliation(s)
- Ivana R. Sequeira
- Human Nutrition Unit, School of Biological Sciences, Faculty of Science, University of Auckland, Auckland, New Zealand
- High-Value Nutrition National Science Challenge, Auckland, New Zealand
- * E-mail:
| | - Wilson Yip
- Human Nutrition Unit, School of Biological Sciences, Faculty of Science, University of Auckland, Auckland, New Zealand
- High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Louise W. Lu
- Human Nutrition Unit, School of Biological Sciences, Faculty of Science, University of Auckland, Auckland, New Zealand
- High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Yannan Jiang
- Department of Statistics, Faculty of Science, University of Auckland, Auckland, New Zealand
| | - Rinki Murphy
- High-Value Nutrition National Science Challenge, Auckland, New Zealand
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Auckland District Health Board, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Lindsay D. Plank
- Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Garth J. S. Cooper
- Division of Cardiovascular Sciences, Centre for Advanced Discovery and Experimental Therapeutics (CADET), Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
- School of Biological Sciences, Faculty of Science, University of Auckland, Auckland, New Zealand
- Division of Medical Sciences, Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Carl N. Peters
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Waitemata District Health Board, Auckland, New Zealand
| | - Benjamin S. Aribsala
- Newcastle Magnetic Resonance Centre, Translational and Clinical Research Institute, Faculty of Medical Science, Newcastle University, Newcastle Upon Tyne, United Kingdom
- Department of Computer Science, Faculty of Science, Lagos State University, Lagos, Nigeria
| | - Kieren G. Hollingsworth
- Newcastle Magnetic Resonance Centre, Translational and Clinical Research Institute, Faculty of Medical Science, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Sally D. Poppitt
- Human Nutrition Unit, School of Biological Sciences, Faculty of Science, University of Auckland, Auckland, New Zealand
- High-Value Nutrition National Science Challenge, Auckland, New Zealand
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Riddet Centre of Research Excellence (CoRE) for Food and Nutrition, Palmerston North, New Zealand
| |
Collapse
|
4
|
Dey S, Murmu N, Mondal T, Saha I, Chatterjee S, Manna R, Haldar S, Dash SK, Sarkar TR, Giri B. Multifaceted entrancing role of glucose and its analogue, 2-deoxy-D-glucose in cancer cell proliferation, inflammation, and virus infection. Biomed Pharmacother 2022; 156:113801. [DOI: 10.1016/j.biopha.2022.113801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/29/2022] [Accepted: 10/02/2022] [Indexed: 11/30/2022] Open
|
5
|
Dietrich JW, Dasgupta R, Anoop S, Jebasingh F, Kurian ME, Inbakumari M, Boehm BO, Thomas N. SPINA Carb: a simple mathematical model supporting fast in-vivo estimation of insulin sensitivity and beta cell function. Sci Rep 2022; 12:17659. [PMID: 36271244 PMCID: PMC9587026 DOI: 10.1038/s41598-022-22531-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 10/17/2022] [Indexed: 01/18/2023] Open
Abstract
Modelling insulin-glucose homeostasis may provide novel functional insights. In particular, simple models are clinically useful if they yield diagnostic methods. Examples include the homeostasis model assessment (HOMA) and the quantitative insulin sensitivity check index (QUICKI). However, limitations of these approaches have been criticised. Moreover, recent advances in physiological and biochemical research prompt further refinement in this area. We have developed a nonlinear model based on fundamental physiological motifs, including saturation kinetics, non-competitive inhibition, and pharmacokinetics. This model explains the evolution of insulin and glucose concentrations from perturbation to steady-state. Additionally, it lays the foundation of a structure parameter inference approach (SPINA), providing novel biomarkers of carbohydrate homeostasis, namely the secretory capacity of beta-cells (SPINA-GBeta) and insulin receptor gain (SPINA-GR). These markers correlate with central parameters of glucose metabolism, including average glucose infusion rate in hyperinsulinemic glucose clamp studies, response to oral glucose tolerance testing and HbA1c. Moreover, they mirror multiple measures of body composition. Compared to normal controls, SPINA-GR is significantly reduced in subjects with diabetes and prediabetes. The new model explains important physiological phenomena of insulin-glucose homeostasis. Clinical validation suggests that it may provide an efficient biomarker panel for screening purposes and clinical research.
Collapse
Affiliation(s)
- Johannes W. Dietrich
- grid.5570.70000 0004 0490 981XDiabetes, Endocrinology and Metabolism Section, Department of Internal Medicine I, St. Josef Hospital, Ruhr University Bochum, NRW, Gudrunstr. 56, 44791 Bochum, Germany ,Diabetes Centre Bochum-Hattingen, St. Elisabeth-Hospital Blankenstein, Im Vogelsang 5-11, 45527 Hattingen, NRW Germany ,grid.5570.70000 0004 0490 981XCentre for Rare Endocrine Diseases, Ruhr Centre for Rare Diseases (CeSER), Ruhr University Bochum and Witten/Herdecke University, Alexandrinenstr. 5, 44791 Bochum, NRW Germany ,Centre for Diabetes Technology, Catholic Hospitals Bochum, Gudrunstr. 56, 44791 Bochum, NRW, Germany
| | - Riddhi Dasgupta
- grid.11586.3b0000 0004 1767 8969Department of Endocrinology, Diabetes and Metabolism, Christian Medical College, Vellore, 632004 India
| | - Shajith Anoop
- grid.11586.3b0000 0004 1767 8969Department of Endocrinology, Diabetes and Metabolism, Christian Medical College, Vellore, 632004 India
| | - Felix Jebasingh
- grid.11586.3b0000 0004 1767 8969Department of Endocrinology, Diabetes and Metabolism, Christian Medical College, Vellore, 632004 India
| | - Mathews E. Kurian
- grid.11586.3b0000 0004 1767 8969Department of Endocrinology, Diabetes and Metabolism, Christian Medical College, Vellore, 632004 India
| | - Mercy Inbakumari
- grid.11586.3b0000 0004 1767 8969Department of Endocrinology, Diabetes and Metabolism, Christian Medical College, Vellore, 632004 India
| | - Bernhard O. Boehm
- grid.59025.3b0000 0001 2224 0361Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore, 308232 Singapore ,grid.6582.90000 0004 1936 9748Department of Internal Medicine I, Ulm University Medical Centre, Ulm University, 89070 Ulm, Germany ,grid.240988.f0000 0001 0298 8161Department of Endocrinology, Tan Tock Seng Hospital, Singapore, Singapore
| | - Nihal Thomas
- grid.11586.3b0000 0004 1767 8969Department of Endocrinology, Diabetes and Metabolism, Christian Medical College, Vellore, 632004 India
| |
Collapse
|
6
|
Bonora E, Trombetta M, Dauriz M, Brangani C, Cacciatori V, Negri C, Pichiri I, Stoico V, Rinaldi E, Da Prato G, Boselli ML, Santi L, Moschetta F, Zardini M, Bonadonna RC. Insulin resistance and beta-cell dysfunction in newly diagnosed type 2 diabetes: Expression, aggregation and predominance. Verona Newly Diagnosed Type 2 Diabetes Study 10. Diabetes Metab Res Rev 2022; 38:e3558. [PMID: 35717608 PMCID: PMC9786655 DOI: 10.1002/dmrr.3558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 05/10/2022] [Accepted: 05/23/2022] [Indexed: 12/30/2022]
Abstract
AIMS We investigated quantitative expression, mutual aggregation and relation with hyperglycemia of insulin resistance (IR) and beta-cell dysfunction (BCD) in newly diagnosed type 2 diabetes. METHODS We assessed IR with euglycemic hyperinsulinemic clamp and BCD with modelled glucose/C-peptide response to oral glucose in 729 mostly drug-naïve patients. We measured glycated hemoglobin, pre-prandial, post-prandial and meal-related excursion of blood glucose. RESULTS IR was found in 87.8% [95% confidence intervals 85.4-90.2] and BCD in 90.0% [87.8-92.2] of subjects, ranging from mild to moderate or severe. Approximately 20% of subjects had solely one defect: BCD 10.8% [8.6-13.1] or IR 8.6% [6.6-10.7]. Insulin resistance and BCD aggregated in most subjects (79.1% [76.2-82.1]). We arbitrarily set nine possible combinations of mild, moderate or severe IR and mild, moderate or severe BCD, finding that each had a similar frequency (∼10%). In multiple regression analyses parameters of glucose control were related more strongly with BCD than with IR. CONCLUSIONS In newly-diagnosed type 2 diabetes, IR and BCD are very common with a wide range of expression but no specific pattern of aggregation. Beta-cell dysfunction is likely to play a greater quantitative role than IR in causing/sustaining hyperglycemia in newly-diagnosed type 2 diabetes.
Collapse
Affiliation(s)
- Enzo Bonora
- Division of Endocrinology, Diabetes and Metabolic Diseases, Department of MedicineUniversity and Hospital Trust of VeronaVeronaItaly
| | - Maddalena Trombetta
- Division of Endocrinology, Diabetes and Metabolic Diseases, Department of MedicineUniversity and Hospital Trust of VeronaVeronaItaly
| | - Marco Dauriz
- Division of Endocrinology, Diabetes and Metabolic Diseases, Department of MedicineUniversity and Hospital Trust of VeronaVeronaItaly
| | - Corinna Brangani
- Division of Endocrinology, Diabetes and Metabolic Diseases, Department of MedicineUniversity and Hospital Trust of VeronaVeronaItaly
| | - Vittorio Cacciatori
- Division of Endocrinology, Diabetes and Metabolic Diseases, Department of MedicineUniversity and Hospital Trust of VeronaVeronaItaly
| | - Carlo Negri
- Division of Endocrinology, Diabetes and Metabolic Diseases, Department of MedicineUniversity and Hospital Trust of VeronaVeronaItaly
| | - Isabella Pichiri
- Division of Endocrinology, Diabetes and Metabolic Diseases, Department of MedicineUniversity and Hospital Trust of VeronaVeronaItaly
| | - Vincenzo Stoico
- Division of Endocrinology, Diabetes and Metabolic Diseases, Department of MedicineUniversity and Hospital Trust of VeronaVeronaItaly
| | - Elisabetta Rinaldi
- Division of Endocrinology, Diabetes and Metabolic Diseases, Department of MedicineUniversity and Hospital Trust of VeronaVeronaItaly
| | - Giuliana Da Prato
- Division of Endocrinology, Diabetes and Metabolic Diseases, Department of MedicineUniversity and Hospital Trust of VeronaVeronaItaly
| | - Maria Linda Boselli
- Division of Endocrinology, Diabetes and Metabolic Diseases, Department of MedicineUniversity and Hospital Trust of VeronaVeronaItaly
| | - Lorenza Santi
- Division of Endocrinology, Diabetes and Metabolic Diseases, Department of MedicineUniversity and Hospital Trust of VeronaVeronaItaly
| | - Federica Moschetta
- Division of Endocrinology, Diabetes and Metabolic Diseases, Department of MedicineUniversity and Hospital Trust of VeronaVeronaItaly
| | - Monica Zardini
- Division of Endocrinology, Diabetes and Metabolic Diseases, Department of MedicineUniversity and Hospital Trust of VeronaVeronaItaly
| | - Riccardo C. Bonadonna
- Department of Medicine and SurgeryUniversity of Parma, and Division of Endocrinology and Metabolic DiseasesAzienda Ospedaliero‐Universitaria di ParmaParmaItaly
| |
Collapse
|
7
|
Koh HCE, Patterson BW, Reeds DN, Mittendorfer B. Insulin sensitivity and kinetics in African American and White people with obesity: Insights from different study protocols. Obesity (Silver Spring) 2022; 30:655-665. [PMID: 35083870 PMCID: PMC8866210 DOI: 10.1002/oby.23363] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/28/2021] [Accepted: 12/02/2021] [Indexed: 01/03/2023]
Abstract
OBJECTIVE Studies that used an intravenous glucose tolerance test (IVGTT) have suggested that race is an important modulator of insulin sensitivity, β-cell function, and insulin clearance. However, the validity of the IVGTT has been challenged. METHODS This study assessed insulin sensitivity and insulin kinetics in non-Hispanic White (NHW, n = 29) and African American (AA, n = 14) people with obesity by using a hyperinsulinemic-euglycemic pancreatic clamp with glucose tracer infusion, an oral glucose tolerance test (OGTT), and an IVGTT. RESULTS Hepatic insulin sensitivity was better in AA participants than in NHW participants. Muscle insulin sensitivity, insulin secretion in relation to plasma glucose during the OGTT, and insulin clearance during basal conditions during the hyperinsulinemic-euglycemic pancreatic clamp and during the OGTT were not different between AA participants and NHW participants. The acute insulin response to the large glucose bolus administered during the IVGTT was double in AA participants compared with NHW participants because of increased insulin secretion and reduced insulin clearance. CONCLUSIONS AA individuals are not more insulin resistant than NHW individuals, and the β-cell response to glucose ingestion and postprandial insulin clearance are not different between AA individuals and NHW individuals. However, AA individuals have greater insulin secretory capacity and reduced insulin clearance capacity than NHW individuals and might be susceptible to hyperinsulinemia after consuming very large amounts of glucose.
Collapse
Affiliation(s)
- Han-Chow E Koh
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Bruce W Patterson
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Dominic N Reeds
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Bettina Mittendorfer
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
8
|
Bizzotto R, Tricò D, Natali A, Gastaldelli A, Muscelli E, De Fronzo RA, Arslanian S, Ferrannini E, Mari A. New Insights on the Interactions Between Insulin Clearance and the Main Glucose Homeostasis Mechanisms. Diabetes Care 2021; 44:2115-2123. [PMID: 34362813 DOI: 10.2337/dc21-0545] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 06/14/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Endogenous insulin clearance (EIC) is physiologically reduced at increasing insulin secretion rate (ISR). Computing EIC at the prevailing ISR does not distinguish the effects of hypersecretion from those of other mechanisms of glucose homeostasis. We aimed to measure EIC in standardized ISR conditions (i.e., at fixed ISR levels) and to analyze its associations with relevant physiologic factors. RESEARCH DESIGN AND METHODS We estimated standardized EIC (EICISR) by mathematical modeling in nine different studies with insulin and glucose infusions (N = 2,067). EICISR association with various traits was analyzed by stepwise multivariable regression in studies with both euglycemic clamp and oral glucose tolerance test (OGTT) (N = 1,410). We also tested whether oral glucose ingestion, as opposed to intravenous infusion, has an independent effect on EIC (N = 1,555). RESULTS Insulin sensitivity (as M/I from the euglycemic clamp) is the strongest determinant of EICISR, approximately four times more influential than insulin resistance-related hypersecretion. EICISR independently associates positively with M/I, fasting and mean OGTT glucose or type 2 diabetes, and β-cell glucose sensitivity and negatively with African American or Hispanic race, female sex, and female age. With oral glucose ingestion, an ISR-independent ∼10% EIC reduction is necessary to explain the observed insulin concentration profiles. CONCLUSIONS Based on EICISR, we posit the existence of two adaptive processes involving insulin clearance: the first reduces EICISR with insulin resistance (not with higher BMI per se) and is more relevant than the concomitant hypersecretion; the second reduces EICISR with β-cell dysfunction. These processes are dysregulated in type 2 diabetes. Finally, oral glucose ingestion per se reduces insulin clearance.
Collapse
Affiliation(s)
| | - Domenico Tricò
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Andrea Natali
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Elza Muscelli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Ralph A De Fronzo
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Silva Arslanian
- Center for Pediatric Research in Obesity and Metabolism, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA
| | | | | |
Collapse
|
9
|
Caturano A, Galiero R, Pafundi PC, Cesaro A, Vetrano E, Palmiero G, Rinaldi L, Salvatore T, Marfella R, Sardu C, Moscarella E, Gragnano F, Calabrò P, Sasso FC. Does a strict glycemic control during acute coronary syndrome play a cardioprotective effect? Pathophysiology and clinical evidence. Diabetes Res Clin Pract 2021; 178:108959. [PMID: 34280467 DOI: 10.1016/j.diabres.2021.108959] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/13/2021] [Accepted: 07/14/2021] [Indexed: 01/08/2023]
Abstract
A hyperglycemic state, also in non-diabetic subjects, may be associated with acute coronary syndrome (ACS). Aim of this review is to describe the pathophysiologic association between ACS and hyperglycemic state, the protective mechanisms of a tight glycaemic control in ACS on CV outcomes, and the supporting clinical evidence. Several mechanisms may be responsible of a poor CV outcome in subjects with hyperglycemia during ACS. Endothelial NAPDH oxidase-2 (NOX2) activation in response to high glucose alters the balance between Raf/MAPK-dependent vasoconstriction and PI3K/Akt-dependent vasodilation in favour of constriction. Hyperglycaemia induces an overproduction of superoxide by the mitochondrial electron transport chain through different molecular mechanisms. Moreover, hyperglycaemia increases the size of the infarct by causing myocardial cell death through apoptosis and reducing the collateral blood flow. High FFA concentrations lead to toxicity mechanisms in acutely ischemic myocardium. On the other hand, a tight glycaemic control in ACS exerts a cardioprotective action by anti-inflammatory and anti-apoptotic mechanisms, anti-oxidative stress, endothelium protection, FFA reduction, anti-glucotoxic effect, IR and cardiac fuel metabolisms improvement, heart stem cells protection and reduced activation of adrenergic system. Unfortunately, the clinical studies supporting the above pathophysiological background are few and sometimes controversial, more likely due the risk of hypoglycemia linked to the insulin therapy generally used during ACS. Intriguingly, GLP-1 RA and SGLT2i, demonstrated highly effective in the cardiovascular prevention in high-risk subjects without the risk of hypoglycemia, might keep this cardioprotective effect even in acute conditions such as ASC.
Collapse
Affiliation(s)
- Alfredo Caturano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy
| | - Raffaele Galiero
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy
| | - Pia Clara Pafundi
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy
| | - Arturo Cesaro
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", 80131 Naples, Italy; Division of Cardiology, A.O.R.N. "Sant'Anna & San Sebastiano", 81100 Caserta, Italy
| | - Erica Vetrano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy
| | - Giuseppe Palmiero
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", 80131 Naples, Italy
| | - Luca Rinaldi
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy
| | - Teresa Salvatore
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via De Crecchio 7, I-80138 Naples, Italy
| | - Raffaele Marfella
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy
| | - Celestino Sardu
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy
| | - Elisabetta Moscarella
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", 80131 Naples, Italy; Division of Cardiology, A.O.R.N. "Sant'Anna & San Sebastiano", 81100 Caserta, Italy
| | - Felice Gragnano
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", 80131 Naples, Italy; Division of Cardiology, A.O.R.N. "Sant'Anna & San Sebastiano", 81100 Caserta, Italy
| | - Paolo Calabrò
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", 80131 Naples, Italy; Division of Cardiology, A.O.R.N. "Sant'Anna & San Sebastiano", 81100 Caserta, Italy
| | - Ferdinando Carlo Sasso
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy.
| |
Collapse
|
10
|
Mengistu Y, Dedefo G, Arkew M, Asefa G, Jebessa G, Atnafu A, Ataro Z, Kinde S. Effect of Regular Khat Chewing on Serum Fasting Sugar Level in Diabetic patients versus Healthy Individuals; A comparative study. Nutr Metab Insights 2021; 14:11786388211035220. [PMID: 34376995 PMCID: PMC8320567 DOI: 10.1177/11786388211035220] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 07/08/2021] [Indexed: 11/17/2022] Open
Abstract
Background Khat chewing is a long standing social-cultural habit in several countries. Even though many people chew khat simply for its pleasurable and stimulatory effect, evidence showed widely-held belief among khat chewers in Ethiopia and other part of the world that khat helps to lower blood glucose while some studies are contradicted on the effect of khat. There is limited data about khat's effect on blood glucose especially in our setting, Harar estern Ethiopia. Objective Primarily the present study aims to compare fasting blood sugar level among khat chewer diabetic and healthy individuals, and to asses risk factors associated with poor glycemic control in diabetic subjects. Method A cross-sectional study included 200 confirmed diabetic and healthy subjects. Fasting blood sugar was determined by enzymatic method glucose oxidase and glucose hexokinase. Glycemic control was also determined for diabetic subjects based on the last 2-month diabetic clinic visits and current measurement. Result (Median ± IQR [interquartile range]) fasting blood sugar difference among Khat chewer and non khat chewer were 159 ± 83 mg/dl and 202 ± 79 mg/dl respectively in diabetic subjects when tested by glucose oxidase. Similarly, in healthy non khat chewer and khat chewer, khat chewers has lower (Median ± IQR) fasting blood glucose level 82 ± 18 mg/dl than non khat chewers 94 ± 13 mg/dl when tested by glucose oxidase. Regarding risk factors associated with poor glycemic control in diabetic subjects, positive parental diabetes history, insulin medication, being overweight, obese were significantly associated with poor glycemic control. Conclusion There was significant effect of khat on median FBS among khat chewers in diabetic and healthy individuals. And the proportion of glycemic control was high among diabetic subjects. Recommendation Health care professional and patients should manage the risk factors to delay disease progression and restrain the damage. More studies should be conducted in randomized control trial manner to further elucidate khat effect on blood sugar level so that the actual effect of khat can be identified unlike in cross sectional where there may not be strong causal relationship.
Collapse
Affiliation(s)
- Yordanos Mengistu
- Department of Medical Laboratory, School of Allied Health Sciences, Health Science College, Addis Ababa University, Ethiopia
| | - Gobena Dedefo
- Department of Medical Laboratory, School of Allied Health Sciences, Health Science College, Addis Ababa University, Ethiopia
| | - Mesay Arkew
- Department of Medical Laboratory, College of Health and Medical Science, Haramaya University, Ethiopia
| | - Gebeyehu Asefa
- Armauer Hanson Research institute, Addis Ababa, Ethiopia
| | - Gutema Jebessa
- Armauer Hanson Research institute, Addis Ababa, Ethiopia
| | - Abay Atnafu
- Armauer Hanson Research institute, Addis Ababa, Ethiopia
| | - Zerihun Ataro
- Department of Medical Laboratory, College of Health and Medical Science, Haramaya University, Ethiopia
| | - Samuel Kinde
- Department of Medical Laboratory, School of Allied Health Sciences, Health Science College, Addis Ababa University, Ethiopia
| |
Collapse
|
11
|
Henquin JC. Glucose-induced insulin secretion in isolated human islets: Does it truly reflect β-cell function in vivo? Mol Metab 2021; 48:101212. [PMID: 33737253 PMCID: PMC8065218 DOI: 10.1016/j.molmet.2021.101212] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/03/2021] [Accepted: 03/09/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Diabetes always involves variable degrees of β-cell demise and malfunction leading to insufficient insulin secretion. Besides clinical investigations, many research projects used rodent islets to study various facets of β-cell pathophysiology. Their important contributions laid the foundations of steadily increasing numbers of experimental studies resorting to isolated human islets. SCOPE OF REVIEW This review, based on an analysis of data published over 60 years of clinical investigations and results of more recent studies in isolated islets, addresses a question of translational nature. Does the information obtained in vitro with human islets fit with our knowledge of insulin secretion in man? The aims are not to discuss specificities of pathways controlling secretion but to compare qualitative and quantitative features of glucose-induced insulin secretion in isolated human islets and in living human subjects. MAJOR CONCLUSIONS Much of the information gathered in vitro can reliably be translated to the in vivo situation. There is a fairly good, though not complete, qualitative and quantitative coherence between insulin secretion rates measured in vivo and in vitro during stimulation with physiological glucose concentrations, but the concordance fades out under extreme conditions. Perplexing discrepancies also exist between insulin secretion in subjects with Type 2 diabetes and their islets studied in vitro, in particular concerning the kinetics. Future projects should ascertain that the experimental conditions are close to physiological and do not alter the function of normal and diabetic islets.
Collapse
Affiliation(s)
- Jean-Claude Henquin
- Unit of Endocrinology and Metabolism, Faculty of Medicine, University of Louvain, Brussels, Belgium.
| |
Collapse
|
12
|
Abriz AE, Rahbarghazi R, Nourazarian A, Avci ÇB, Mahboob SA, Rahnema M, Araghi A, Heidarzadeh M. Effect of docosahexaenoic acid plus insulin on atherosclerotic human endothelial cells. JOURNAL OF INFLAMMATION-LONDON 2021; 18:10. [PMID: 33602249 PMCID: PMC7890865 DOI: 10.1186/s12950-021-00277-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 02/08/2021] [Indexed: 12/25/2022]
Abstract
Background Atherosclerosis is touted as one of the most critical consequences of diabetes mellitus indicated by local inflammation of endothelial cells. The Effect of Omega 3 fatty acids, mainly docosahexaenoic acid (DHA), has been investigated in cells after exposure to high doses of lipids. The current experiment aimed to address the modulatory effects of docosahexaenoic acid and insulin in palmitic-treated human endothelial cells. Methods Human umbilical vein endothelial cells were treated with 1 mM palmitic acid, 50 μM insulin, 50 μM docosahexaenoic acid, and their combination for 48 h. Cell survival rate and apoptosis were measured using MTT and flow cytometry assays. The Griess assay detected NO levels. Protein levels of TNF-α, IL-6, and NF-κB were studied using ELISA and immunofluorescence imaging. The expression of genes participating in atherosclerosis was monitored using PCR array analysis. Results Oil Red O staining showed the inhibitory effect of DHA and insulin to reduce the intracellular accumulation of palmitic acid. Both DHA and Insulin blunted palmitic acid detrimental effects on HUVECs indicated by an increased survival rate (p < 0.05). The percent of apoptotic cells was decreased in palmitic-treated cells received insulin and DHA compared to palmitic-treated group (p < 0.05). Based on our data, DHA and Insulin diminished the production of all inflammatory cytokines, TNF-α, IL-6, and NF-κB, in palmitic-treated cells (p < 0.05). Similar to these data, NO production was also decreased in all groups treated with insulin and DHA compared to the palmitic-treated cells (p < 0.05). PCR array analysis revealed the modulatory effect of DHA and insulin on the expression of atherosclerosis-related genes pre-treated with palmitic acid compared to the control group (p < 0.05). Conclusion DHA and Insulin could alter the dynamic growth and dysfunctional activity of human endothelial cells after treatment with palmitic acid. Taken together, Omega 3 fatty acids, along with insulin, could dictate specific cell behavior in endothelial cells in vitro.
Collapse
Affiliation(s)
- Aysan Eslami Abriz
- Department of Biochemistry, Higher Education Institute of Rab-Rashid, Tabriz, Iran.,Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Nourazarian
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Golgasht St, Tabriz, 51666-16471, Iran.
| | - Çıgır Biray Avci
- Department of Medical Biology, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Soltan Ali Mahboob
- Department of Biochemistry, Higher Education Institute of Rab-Rashid, Tabriz, Iran
| | - Maryam Rahnema
- Department of Biochemistry, Higher Education Institute of Rab-Rashid, Tabriz, Iran.,Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Atefeh Araghi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Amol University of Special Modern Technologies, Amol, Iran
| | - Morteza Heidarzadeh
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
13
|
Al-Disi D, Ansari MGA, Sabico S, Wani K, Hussain SD, Elshafie MM, McTernan P, Al-Daghri NM. High glucose load and endotoxemia among overweight and obese Arab women with and without diabetes: An observational study. Medicine (Baltimore) 2020; 99:e23211. [PMID: 33181703 PMCID: PMC7668447 DOI: 10.1097/md.0000000000023211] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Dietary intake influences gut microbiota activity. Nevertheless, there is a lack of evidence available that illustrates the acute effects of high glucose meal on metabolic endotoxemia. The present study assessed the acute impact of high glucose meal on endotoxemia and other clinical parameters in Saudi females with varying degrees of glycemia.The subjects were 64 consenting pre-menopausal women, grouped into 3: control [n = 14 lean, non-T2DM, BMI = 22.2 ± 2.2 kg/m]; overweight [n = 16, non-T2DM, BMI = 28.5 ± 1.5 kg/m] and T2DM [n = 34, BMI = 35.2 ± 7.7 kg/m]. After an overnight fast, all subjects were given a standardized high-glucose (75 g) meal. Anthropometrics were taken and blood samples were withdrawn at baseline and postprandial (0, 2 and 4-hours), serum glucose, endotoxin and lipid profile were quantified.At baseline, total cholesterol, LDL-cholesterol, triglycerides and serum glucose levels were significantly higher (P values <.01) whereas significantly lower HDL-cholesterol levels (P < .01) were observed in T2DM subjects compared to other groups. Baseline endotoxin levels were highest in the overweight group (3.2 ± 1.1 mmol/L) as compared to control (2.0 ± 0.5 mmol/L) and T2DM (2.7 ± 1.2 mmol/L) (P = .046). HDL-cholesterol, LDL-cholesterol and triglycerides, significantly decreased in the T2DM group after 2 hours (P values <.05), whereas unremarkable changes observed in other groups. Lastly, endotoxin levels significantly increased only in the overweight group (3.2 ± 1.1 vs 4.2 ± 1.4 mmol/L; P < .05), 4 hours postprandial.High glucose meal elevates endotoxemia only among overweight subjects and impairs dysbiosis.
Collapse
Affiliation(s)
- Dara Al-Disi
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University
| | | | - Shaun Sabico
- Riyadh Biochemistry Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Kaiser Wani
- Riyadh Biochemistry Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Syed Danish Hussain
- Riyadh Biochemistry Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mona M. Elshafie
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University
| | - Philip McTernan
- School of Science and Technology, Department of Biosciences, Nottingham Trent University, Nottingham, NG1 8NS, UK
| | - Nasser M. Al-Daghri
- Riyadh Biochemistry Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
14
|
Mengozzi A, Tricò D, Nesti L, Petrie J, Højlund K, Mitrakou A, Krebs M, Mari A, Natali A. Disruption of fasting and post-load glucose homeostasis are largely independent and sustained by distinct and early major beta-cell function defects: a cross-sectional and longitudinal analysis of the Relationship between Insulin Sensitivity and Cardiovascular risk (RISC) study cohort. Metabolism 2020; 105:154185. [PMID: 32061908 DOI: 10.1016/j.metabol.2020.154185] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 02/10/2020] [Accepted: 02/12/2020] [Indexed: 01/26/2023]
Abstract
BACKGROUND/AIMS Uncertainty still exists on the earliest beta-cell defects at the bases of the type 2 diabetes. We assume that this depends on the inaccurate distinction between fasting and post-load glucose homeostasis and aim at providing a description of major beta-cell functions across the full physiologic spectrum of each condition. METHODS In 1320 non-diabetic individuals we performed an OGTT with insulin secretion modeling and a euglycemic insulin clamp, coupled in subgroups to glucose tracers and IVGTT; 1038 subjects underwent another OGTT after 3.5 years. Post-load glucose homeostasis was defined as mean plasma glucose above fasting levels (δOGTT). The analysis was performed by two-way ANCOVA. RESULTS Fasting plasma glucose (FPG) and δOGTT were weakly related variables (stβ = 0.12) as were their changes over time (r = -0.08). Disruption of FPG control was associated with an isolated and progressive decline (approaching 60%) of the sensitivity of the beta-cell to glucose values within the normal fasting range. Disruption of post-load glucose control was characterized by a progressive decline (approaching 60%) of the slope of the full beta-cell vs glucose dose-response curve and an early minor (30%) decline of potentiation. The acute dynamic beta-cell responses, neither per se nor in relation to the degree of insulin resistance appeared to play a relevant role in disruption of fasting or post-load homeostasis. Follow-up data qualitatively and quantitatively confirmed the results of the cross-sectional analysis. CONCLUSION In normal subjects fasting and post-load glucose homeostasis are largely independent, and their disruption is sustained by different and specific beta-cell defects.
Collapse
Affiliation(s)
- Alessandro Mengozzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.
| | - Domenico Tricò
- Department of Surgical, Medical, Molecular and Critical Area Pathology, University of Pisa, Italy
| | - Lorenzo Nesti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - John Petrie
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Kurt Højlund
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark
| | - Asimina Mitrakou
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Alexandra Hospital, Athens, Greece
| | - Michael Krebs
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Andrea Mari
- Institute of Neuroscience, National Research Council, Padua, Italy
| | - Andrea Natali
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
15
|
Zhyzhneuskaya SV, Al-Mrabeh A, Peters C, Barnes A, Aribisala B, Hollingsworth KG, McConnachie A, Sattar N, Lean MEJ, Taylor R. Time Course of Normalization of Functional β-Cell Capacity in the Diabetes Remission Clinical Trial After Weight Loss in Type 2 Diabetes. Diabetes Care 2020; 43:813-820. [PMID: 32060017 DOI: 10.2337/dc19-0371] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 12/29/2019] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To assess functional β-cell capacity in type 2 diabetes during 2 years of remission induced by dietary weight loss. RESEARCH DESIGN AND METHODS A Stepped Insulin Secretion Test with Arginine was used to quantify functional β-cell capacity by hyperglycemia and arginine stimulation. Thirty-nine of 57 participants initially achieved remission (HbA1c <6.5% [<48 mmol/mol] and fasting plasma glucose <7 mmol/L on no antidiabetic drug therapy) with a 16.4 ± 7.7 kg weight loss and were followed up with supportive advice on avoidance of weight regain. At 2 years, 20 participants remained in remission in the study. A nondiabetic control (NDC) group, matched for age, sex, and weight after weight loss with the intervention group, was studied once. RESULTS During remission, median (interquartile range) maximal rate of insulin secretion increased from 581 (480-811) pmol/min/m2 at baseline to 736 (542-998) pmol/min/m2 at 5 months, 942 (565-1,240) pmol/min/m2 at 12 months (P = 0.028 from baseline), and 936 (635-1,435) pmol/min/m2 at 24 months (P = 0.023 from baseline; n = 20 of 39 of those initially in remission). This was comparable to the NDC group (1,016 [857-1,507] pmol/min/m2) by 12 (P = 0.064) and 24 (P = 0.244) months. Median first-phase insulin response increased from baseline to 5 months (42 [4-67] to 107 [59-163] pmol/min/m2; P < 0.0001) and then remained stable at 12 and 24 months (110 [59-201] and 125 [65-166] pmol/min/m2, respectively; P < 0.0001 vs. baseline) but lower than that of the NDC group (250 [226-429] pmol/min/m2; P < 0.0001). CONCLUSIONS A gradual increase in assessed functional β-cell capacity occurred after weight loss, becoming similar to that of NDC group participants by 12 months. This result was unchanged at 2 years with continuing remission of type 2 diabetes.
Collapse
Affiliation(s)
- Sviatlana V Zhyzhneuskaya
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, U.K
| | - Ahmad Al-Mrabeh
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, U.K
| | - Carl Peters
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, U.K
| | - Alison Barnes
- Human Nutrition Research Centre, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, U.K
| | | | - Kieren G Hollingsworth
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, U.K
| | - Alex McConnachie
- Robertson Centre for Biostatistics, University of Glasgow, Glasgow, U.K
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, U.K
| | - Michael E J Lean
- School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, U.K
| | - Roy Taylor
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, U.K.
| |
Collapse
|
16
|
Acute glucose load induced islet β cells dysfunction in TLR4 dependent manner in male mice. Biochem Biophys Res Commun 2020; 524:205-210. [PMID: 31983426 DOI: 10.1016/j.bbrc.2020.01.059] [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: 12/15/2019] [Accepted: 01/11/2020] [Indexed: 11/21/2022]
Abstract
Recent studies highlighted the significance of chronic inflammation, which is mediated in part by toll-like receptors 4 (TLR4), in islet β cell dysfunction by high-glucose exposure. However, about it is unclear whether islet β cell dysfunction in response to high glucose is associated with TLR4. This investigation was designed to address the effect of TLR4 deficiency on insulin secretion in mice in response to acute intravenous glucose load. Hyperglycemic clamp was used to impair insulin secretion, and intraperitoneal glucose tolerance test was carried out to analyze insulin secretion function of islet β cells. Our results showed that TLR4 deficiency repressed insulin secretion impairment in response to acute intravenous glucose load. Compared to wild-type mice, TLR4-/- mice did not exhibit increase of IL-1β and TNF-α level in plasma and pancreatic tissue in response to acute intravenous load of high glucose. However, recombinant IL-1β or TNF-α administration restored insulin secretion impairment induced by high glucose in TLR4-/- mice. Taken together, our results demonstrated that TLR4 activation and subsequent IL-1β and TNF-α production contribute to islet β cell dysfunction in mice in response to acute intravenous load of high glucose, which may provide a theoretical basis for diabetes complication improvement by physical exercise.
Collapse
|
17
|
Al-Mrabeh A, Zhyzhneuskaya SV, Peters C, Barnes AC, Melhem S, Jesuthasan A, Aribisala B, Hollingsworth KG, Lietz G, Mathers JC, Sattar N, Lean MEJ, Taylor R. Hepatic Lipoprotein Export and Remission of Human Type 2 Diabetes after Weight Loss. Cell Metab 2020; 31:233-249.e4. [PMID: 31866441 DOI: 10.1016/j.cmet.2019.11.018] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/31/2019] [Accepted: 11/25/2019] [Indexed: 02/07/2023]
Abstract
The role of hepatic lipoprotein metabolism in diet-induced remission of type 2 diabetes is currently unclear. Here, we determined the contributions of hepatic VLDL1-triglyceride production rate and VLDL1-palmitic acid content to changes in intra-pancreatic fat and return of first phase insulin response in a subgroup of the Diabetes Remission Clinical Trial. Liver fat, VLDL1-triglyceride production, and intra-pancreatic fat decreased after weight loss and remained normalized after 24 months of remission. First-phase insulin response remained increased only in those maintaining diabetes remission. Compared with those in remission at 24 months, individuals who relapsed after initial remission had a greater rise in the content of VLDL1-triglyceride and VLDL1-palmitic acid, re-accumulated intra-pancreatic fat, and lost first-phase response by 24 months. Thus, we observed temporal relationships between VLDL1-triglyceride production, hepatic palmitic acid flux, intra-pancreatic fat, and β-cell function. Weight-related disordered fat metabolism appears to drive development and reversal of type 2 diabetes.
Collapse
Affiliation(s)
- Ahmad Al-Mrabeh
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE4 5PL, UK.
| | - Sviatlana V Zhyzhneuskaya
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Carl Peters
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Alison C Barnes
- Human Nutrition Research Centre, Population and Health Sciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Shaden Melhem
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Aaron Jesuthasan
- School of Medical Education, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Benjamin Aribisala
- Computer Science Department, Lagos State University, Lagos PMB 0001, Nigeria
| | - Kieren G Hollingsworth
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Georg Lietz
- Human Nutrition Research Centre, Population and Health Sciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - John C Mathers
- Human Nutrition Research Centre, Population and Health Sciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Science, University of Glasgow, Glasgow G12 8TA, UK
| | - Michael E J Lean
- School of Medicine, Dentistry and Nursing, Glasgow University, Glasgow G31 2ER, UK
| | - Roy Taylor
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE4 5PL, UK.
| |
Collapse
|
18
|
Elhassan SAM, Candasamy M, Ching TS, Heng YK, Bhattamisra SK. Effect of madecassoside and catalpol in amelioration of insulin sensitivity in pancreatic (INS-1E) β-cell line. Nat Prod Res 2019; 35:4627-4631. [PMID: 31797687 DOI: 10.1080/14786419.2019.1696794] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Currently, type 2 diabetes mellitus (T2D) has emerged as global burden disease. Herbal drugs with antidiabetic activities are attracting the attention. Madecassoside and catalpol are herbal compounds having strong antioxidant and glucose lowering activity. Madecassoside and catalpol were investigated for their effect on insulin sensitivity using pancreatic INS-1E cells. Cytotoxicity of these compounds was evaluated by MTT assay. Glucose-stimulated insulin secretion (GSIS) and expression of insulin signalling proteins were studied in presence of madecassoside and catalpol. Results revealed that madecassoside and catalpol enhanced the GSIS without cytotoxic effect. Madecassoside (30 µM) and catalpol (40 µM) increased the insulin secretion in response to high glucose (16.7 mM) stimulation. Subsequently, madecassoside and catalpol showed elevated expression of p-IRS-1, Akt, and p-Akt proteins. Madecassoside and catalpol after 24 h of incubation in pancreatic INS-1E cells with high glucose concentration (30 mM) ameliorated the insulin secretion.
Collapse
Affiliation(s)
| | - Mayuren Candasamy
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Tan Swee Ching
- School of Postgraduate Studies, International Medical University, Kuala Lumpur, Malaysia
| | - Yap Kah Heng
- School of Postgraduate Studies, International Medical University, Kuala Lumpur, Malaysia
| | - Subrat Kumar Bhattamisra
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| |
Collapse
|
19
|
Henquin JC. The challenge of correctly reporting hormones content and secretion in isolated human islets. Mol Metab 2019; 30:230-239. [PMID: 31767174 PMCID: PMC6829677 DOI: 10.1016/j.molmet.2019.10.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 09/28/2019] [Accepted: 10/07/2019] [Indexed: 12/30/2022] Open
Abstract
Background An increased access of research laboratories to isolated human islets has improved our understanding of the biology of the endocrine pancreas and hence the mechanisms causing diabetes. However, in vitro studies of human islets remain technically challenging, and optimal use of such precious material requires a minimum of rigor and coordination to optimize the reliability and share of the information. A detailed report of the demographics of pancreas donors and of the procedures of islet handling after isolation is important but insufficient. Correct characterization of islet basic functions (a token of quality) at the time of experimentation is also crucial. Scope of review I have analyzed the literature reporting measurements of insulin and glucagon in the human pancreas or isolated human islets. The published information is often fragmentary. Elementary features such as islet size, insulin content, or rate of hormone secretion are either unreported or incorrectly reported in many papers. Although internal comparisons between control and test groups may remain valid, comparisons with data from other laboratories are problematic. The drawbacks, pitfalls and errors of common ways of expressing hormone content or secretion rates are discussed and alternatives to harmonize data presentation are proposed. Major Conclusions Greater coherence and rigor in the report of in vitro studies using human islets are necessary to ensure optimal progress in our understanding of the pathogenesis of diabetes.
Collapse
Affiliation(s)
- Jean-Claude Henquin
- Unit of Endocrinology and Metabolism, Faculty of Medicine, University of Louvain, Brussels, Belgium.
| |
Collapse
|
20
|
De Gaetano A, Hardy TA. A novel fast-slow model of diabetes progression: Insights into mechanisms of response to the interventions in the Diabetes Prevention Program. PLoS One 2019; 14:e0222833. [PMID: 31600232 PMCID: PMC6786566 DOI: 10.1371/journal.pone.0222833] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 09/09/2019] [Indexed: 12/22/2022] Open
Abstract
Several models for the long-term development of T2DM already exist, focusing on the dynamics of the interaction between glycemia, insulinemia and β-cell mass. Current models consider representative (fasting or daily average) glycemia and insulinemia as characterizing the compensation state of the subject at some instant in slow time. This implies that only these representative levels can be followed through time and that the role of fast glycemic oscillations is neglected. An improved model (DPM15) for the long-term progression of T2DM is proposed, introducing separate peripheral and hepatic (liver and kidney) insulin actions. The DPM15 model no longer uses near-equilibrium approximation to separate fast and slow time scales, but rather describes, at each step in slow time, a complete day in the life of the virtual subject in fast time. The model can thus represent both fasting and postprandial glycemic levels and describe the effect of interventions acting on insulin-enhanced tissue glucose disposal or on insulin-inhibited hepatic glucose output, as well as on insulin secretion and β-cell replicating ability. The model can simulate long-term variations of commonly used clinical indices (HOMA-B, HOMA-IR, insulinogenic index) as well as of Oral Glucose Tolerance or Euglycemic Hyperinsulinemic Clamp test results. The model has been calibrated against observational data from the Diabetes Prevention Program study: it shows good adaptation to observations as a function of very plausible values of the parameters describing the effect of such interventions as Placebo, Intensive LifeStyle and Metformin administration.
Collapse
Affiliation(s)
- Andrea De Gaetano
- CNR-IASI BioMatLab (Italian National Research Council - Institute of Analysis, Systems and Computer Science - Biomathematics Laboratory), Rome, Italy
| | - Thomas Andrew Hardy
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana, United States of America
| |
Collapse
|
21
|
Meza CA, La Favor JD, Kim DH, Hickner RC. Endothelial Dysfunction: Is There a Hyperglycemia-Induced Imbalance of NOX and NOS? Int J Mol Sci 2019; 20:ijms20153775. [PMID: 31382355 PMCID: PMC6696313 DOI: 10.3390/ijms20153775] [Citation(s) in RCA: 184] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 07/29/2019] [Accepted: 07/31/2019] [Indexed: 02/07/2023] Open
Abstract
NADPH oxidases (NOX) are enzyme complexes that have received much attention as key molecules in the development of vascular dysfunction. NOX have the primary function of generating reactive oxygen species (ROS), and are considered the main source of ROS production in endothelial cells. The endothelium is a thin monolayer that lines the inner surface of blood vessels, acting as a secretory organ to maintain homeostasis of blood flow. The enzymatic production of nitric oxide (NO) by endothelial NO synthase (eNOS) is critical in mediating endothelial function, and oxidative stress can cause dysregulation of eNOS and endothelial dysfunction. Insulin is a stimulus for increases in blood flow and endothelium-dependent vasodilation. However, cardiovascular disease and type 2 diabetes are characterized by poor control of the endothelial cell redox environment, with a shift toward overproduction of ROS by NOX. Studies in models of type 2 diabetes demonstrate that aberrant NOX activation contributes to uncoupling of eNOS and endothelial dysfunction. It is well-established that endothelial dysfunction precedes the onset of cardiovascular disease, therefore NOX are important molecular links between type 2 diabetes and vascular complications. The aim of the current review is to describe the normal, healthy physiological mechanisms involved in endothelial function, and highlight the central role of NOX in mediating endothelial dysfunction when glucose homeostasis is impaired.
Collapse
Affiliation(s)
- Cesar A Meza
- Department of Nutrition, Food & Exercise Sciences, Florida State University, Tallahassee, FL 32306, USA
| | - Justin D La Favor
- Department of Nutrition, Food & Exercise Sciences, Florida State University, Tallahassee, FL 32306, USA
| | - Do-Houn Kim
- Department of Nutrition, Food & Exercise Sciences, Florida State University, Tallahassee, FL 32306, USA
| | - Robert C Hickner
- Department of Nutrition, Food & Exercise Sciences, Florida State University, Tallahassee, FL 32306, USA.
- Institute of Sports Sciences and Medicine, College of Human Sciences, Florida State University, Tallahassee, FL 32306, USA.
- Department of Biokinetics, Exercise and Leisure Sciences, School of Health Sciences, University of KwaZulu-Natal, Westville 4041, South Africa.
| |
Collapse
|
22
|
Lindåse S, Johansson H, Månsby M, Bröjer J. Repeatability of the hyperglycaemic clamp for assessment of β-cell response and insulin sensitivity in horses. Equine Vet J 2019; 52:126-130. [PMID: 30980682 DOI: 10.1111/evj.13119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 04/09/2019] [Indexed: 11/27/2022]
Abstract
BACKGROUND The hyperglycaemic clamp has been used for measurement of insulin sensitivity (IS) but not for measurement of β-cell response in the horse. However, the repeatability of this test has not been reported for horses. OBJECTIVES To determine the repeatability and reliability of measures for IS and β-cell response to glucose using the hyperglycaemic clamp in horses. STUDY DESIGN Repeated measures, longitudinal study. METHODS Six healthy Standardbred mares underwent a 120-min hyperglycaemic clamp on two occasions with a 10-day washout period. Indices of repeatability and reliability were calculated from measures of IS and β-cell response to glucose derived from each hyperglycaemic clamp. RESULTS Measures of β-cell response to glucose, including area under the insulin response curve in the time interval 0-120 min and during steady state (60-120 min), as well as the mean insulin concentration during steady state had coefficient of variations (CV) of 9.5, 10.5 and 9.3 respectively and intraclass correlation coefficients (ICC) of 0.93, 0.93 and 0.95 respectively. The measure of glucose disposal (M) had lower CV (12.4) and ICC (0.71) compared to measure of IS (M/I-index; CV of 17.4 and ICC of 0.89). MAIN LIMITATIONS The study was conducted in a small number of horses. CONCLUSIONS The hyperglycaemic clamp is a diagnostic method for assessing β-cell response to intravenous glucose with high repeatability and reliability. Insulin sensitivity can be determined with the same test but the repeatability for these measurements is lower compared to the measurements for β-cell response.
Collapse
Affiliation(s)
- S Lindåse
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - H Johansson
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - M Månsby
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - J Bröjer
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| |
Collapse
|
23
|
Pedersen MG, Tagliavini A, Henquin JC. Calcium signaling and secretory granule pool dynamics underlie biphasic insulin secretion and its amplification by glucose: experiments and modeling. Am J Physiol Endocrinol Metab 2019; 316:E475-E486. [PMID: 30620637 DOI: 10.1152/ajpendo.00380.2018] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Glucose-stimulated insulin secretion from pancreatic β-cells is controlled by a triggering pathway that culminates in calcium influx and regulated exocytosis of secretory granules, and by a less understood amplifying pathway that augments calcium-induced exocytosis. In response to an abrupt increase in glucose concentration, insulin secretion exhibits a first peak followed by a lower sustained second phase. This biphasic secretion pattern is disturbed in diabetes. It has been attributed to depletion and subsequent refilling of a readily releasable pool of granules or to the phasic cytosolic calcium dynamics induced by glucose. Here, we apply mathematical modeling to experimental data from mouse islets to investigate how calcium and granule pool dynamics interact to control dynamic insulin secretion. Experimental calcium traces are used as inputs in three increasingly complex models of pool dynamics, which are fitted to insulin secretory patterns obtained using a set of protocols of glucose and tolbutamide stimulation. New calcium and secretion data for so-called staircase protocols, in which the glucose concentration is progressively increased, are presented. These data can be reproduced without assuming any heterogeneity in the model, in contrast to previous modeling, because of nontrivial calcium dynamics. We find that amplification by glucose can be explained by increased mobilization and priming of granules. Overall, our results indicate that calcium dynamics contribute substantially to shaping insulin secretion kinetics, which implies that better insight into the events creating phasic calcium changes in human β-cells is needed to understand the cellular mechanisms that disturb biphasic insulin secretion in diabetes.
Collapse
Affiliation(s)
- Morten Gram Pedersen
- Department of Information Engineering, University of Padova , Padova , Italy
- Department of Mathematics "Tullio Levi-Civita, " University of Padova , Padova , Italy
- Padova Neuroscience Center, University of Padova , Padova , Italy
| | - Alessia Tagliavini
- Department of Information Engineering, University of Padova , Padova , Italy
| | - Jean-Claude Henquin
- Unit of Endocrinology and Metabolism, Faculty of Medicine, University of Louvain , Brussels , Belgium
| |
Collapse
|
24
|
De Gaetano A, Gaz C, Panunzi S. Consistency of compact and extended models of glucose-insulin homeostasis: The role of variable pancreatic reserve. PLoS One 2019; 14:e0211331. [PMID: 30768604 PMCID: PMC6377092 DOI: 10.1371/journal.pone.0211331] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 01/11/2019] [Indexed: 01/16/2023] Open
Abstract
Published compact and extended models of the glucose-insulin physiologic control system are compared, in order to understand why a specific functional form of the compact model proved to be necessary for a satisfactory representation of acute perturbation experiments such as the Intra Venous Glucose Tolerance Test (IVGTT). A spectrum of IVGTT’s of virtual subjects ranging from normal to IFG to IGT to frank T2DM were simulated using an extended model incorporating the population-of-controllers paradigm originally hypothesized by Grodsky, and proven to be able to capture a wide array of experimental results from heterogeneous perturbation procedures. The simulated IVGTT’s were then fitted with the Single-Delay Model (SDM), a compact model with only six free parameters, previously shown to be very effective in delivering precise estimates of insulin sensitivity and secretion during an IVGTT. Comparison of the generating, extended-model parameter values with the obtained compact model estimates shows that the functional form of the nonlinear insulin-secretion term, empirically found to be necessary for the compact model to satisfactorily fit clinical observations, captures the pancreatic reserve level of the simulated virtual patients. This result supports the validity of the compact model as a meaningful analysis tool for the clinical assessment of insulin sensitivity.
Collapse
Affiliation(s)
- Andrea De Gaetano
- CNR-IASI BioMatLab, Consiglio Nazionale delle Ricerche, Istituto di Analisi dei Sistemi ed Informatica, Laboratorio di Biomatematica (Italian National Research Council - Institute for System Analysis and Computer Science - Biomathematics Laboratory), UCSC Largo A. Gemelli 8, Rome, Italy
| | - Claudio Gaz
- CNR-IASI BioMatLab, Consiglio Nazionale delle Ricerche, Istituto di Analisi dei Sistemi ed Informatica, Laboratorio di Biomatematica (Italian National Research Council - Institute for System Analysis and Computer Science - Biomathematics Laboratory), UCSC Largo A. Gemelli 8, Rome, Italy
- Sapienza Università di Roma, Dipartimento di Ingegneria Informatica, Automatica e Gestionale (DIAG) (Department of Computer, Control and Management Engineering), Via Ariosto 25, Rome, Italy
- * E-mail: ,
| | - Simona Panunzi
- CNR-IASI BioMatLab, Consiglio Nazionale delle Ricerche, Istituto di Analisi dei Sistemi ed Informatica, Laboratorio di Biomatematica (Italian National Research Council - Institute for System Analysis and Computer Science - Biomathematics Laboratory), UCSC Largo A. Gemelli 8, Rome, Italy
| |
Collapse
|
25
|
Belikov AV. Age-related diseases as vicious cycles. Ageing Res Rev 2019; 49:11-26. [PMID: 30458244 DOI: 10.1016/j.arr.2018.11.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 10/05/2018] [Accepted: 11/15/2018] [Indexed: 02/07/2023]
Abstract
The mortality rates of age-related diseases (ARDs) increase exponentially with age. Processes described by the exponential growth function typically involve a branching chain reaction or, more generally, a positive feedback loop. Here I propose that each ARD is mediated by one or several positive feedback loops (vicious cycles). I then identify critical vicious cycles in five major ARDs: atherosclerosis, hypertension, diabetes, Alzheimer's and Parkinson's. I also propose that the progression of ARDs can be halted by selectively interrupting the vicious cycles and suggest the most promising targets.
Collapse
Affiliation(s)
- Aleksey V Belikov
- Laboratory of Innovative Medicine, School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Institutsky per., 9, 141701 Dolgoprudny, Moscow Region, Russia.
| |
Collapse
|
26
|
Affram KO, Reddy TL, Osei KM. A Rare Case of Thyrotoxic Periodic Paralysis After Epidural Steroid Injection: A Case Report and Literature Review. AMERICAN JOURNAL OF CASE REPORTS 2018; 19:1453-1458. [PMID: 30531678 PMCID: PMC6293861 DOI: 10.12659/ajcr.911270] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Patient: Male, 36 Final Diagnosis: Epidural steroid induced thyrotoxic periodic paralysis Symptoms: Paralysis Medication: — Clinical Procedure: Epidural steroid injection Specialty: Endorinology and Metabolic
Collapse
Affiliation(s)
- Kwame Ofori Affram
- Department of Internal Medicine, Piedmont Athens Regional Medical Center, Athens, GA, USA
| | - Tanya Luke Reddy
- Department of Internal Medicine, Piedmont Athens Regional Medical Center, Athens, GA, USA
| | - Kofi M Osei
- Department of Internal Medicine, Piedmont Athens Regional Medical Center, Athens, GA, USA
| |
Collapse
|
27
|
Taylor R, Al-Mrabeh A, Zhyzhneuskaya S, Peters C, Barnes AC, Aribisala BS, Hollingsworth KG, Mathers JC, Sattar N, Lean MEJ. Remission of Human Type 2 Diabetes Requires Decrease in Liver and Pancreas Fat Content but Is Dependent upon Capacity for β Cell Recovery. Cell Metab 2018; 28:547-556.e3. [PMID: 30078554 DOI: 10.1016/j.cmet.2018.07.003] [Citation(s) in RCA: 242] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 06/12/2018] [Accepted: 07/06/2018] [Indexed: 01/03/2023]
Abstract
The Diabetes Remission Clinical Trial reported return and persistence of non-diabetic blood glucose control in 46% of people with type 2 diabetes of up to 6 years duration. Detailed metabolic studies were performed on a subgroup (intervention, n = 64; control, n = 26). In the intervention group, liver fat content decreased (16.0% ± 1.3% to 3.1% ± 0.5%, p < 0.0001) immediately after weight loss. Similarly, plasma triglyceride and pancreas fat content decreased whether or not glucose control normalized. Recovery of first-phase insulin response (0.04[-0.05-0.32] to 0.11[0.0005-0.51] nmol/min/m2, p < 0.0001) defined those who returned to non-diabetic glucose control and this was durable at 12 months (0.11[0.005-0.81] nmol/min/m2, p = 0.0001). Responders were similar to non-responders at baseline but had shorter diabetes duration (2.7 ± 0.3 versus 3.8 ± 0.4 years; p = 0.02). This study demonstrates that β cell ability to recover long-term function persists after diagnosis, changing the previous paradigm of irreversible loss of β cell function in type 2 diabetes.
Collapse
Affiliation(s)
- Roy Taylor
- Newcastle Magnetic Resonance Centre, Institute of Cellular Medicine, Newcastle University, Campus for Ageing & Vitality, Newcastle upon Tyne NE4 5PL, UK.
| | - Ahmad Al-Mrabeh
- Newcastle Magnetic Resonance Centre, Institute of Cellular Medicine, Newcastle University, Campus for Ageing & Vitality, Newcastle upon Tyne NE4 5PL, UK
| | - Sviatlana Zhyzhneuskaya
- Newcastle Magnetic Resonance Centre, Institute of Cellular Medicine, Newcastle University, Campus for Ageing & Vitality, Newcastle upon Tyne NE4 5PL, UK
| | - Carl Peters
- Newcastle Magnetic Resonance Centre, Institute of Cellular Medicine, Newcastle University, Campus for Ageing & Vitality, Newcastle upon Tyne NE4 5PL, UK
| | - Alison C Barnes
- Human Nutrition Research Centre, Institute of Health & Society, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Benjamin S Aribisala
- Department of Computer Science, Lagos State University, Ojo, Lagos State, Nigeria
| | - Kieren G Hollingsworth
- Newcastle Magnetic Resonance Centre, Institute of Cellular Medicine, Newcastle University, Campus for Ageing & Vitality, Newcastle upon Tyne NE4 5PL, UK
| | - John C Mathers
- Human Nutrition Research Centre, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Science, University of Glasgow, Glasgow G12 8TA, UK
| | - Michael E J Lean
- Human Nutrition, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow G31 2ER, UK
| |
Collapse
|
28
|
Grespan E, Giorgino T, Arslanian S, Natali A, Ferrannini E, Mari A. Defective Amplifying Pathway of β-Cell Secretory Response to Glucose in Type 2 Diabetes: Integrated Modeling of In Vitro and In Vivo Evidence. Diabetes 2018; 67:496-506. [PMID: 29229615 DOI: 10.2337/db17-1039] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 12/05/2017] [Indexed: 11/13/2022]
Abstract
In vivo studies have investigated the role of β-cell dysfunction in type 2 diabetes (T2D), whereas in vitro research on islets has elucidated key mechanisms that control the insulin secretion rate. However, the relevance of the cellular mechanisms identified in vitro (i.e., the triggering and amplifying pathways) has not been established in vivo. Furthermore, the mechanisms underpinning β-cell dysfunction in T2D remain undetermined. We propose a unifying explanation of several characteristic features of insulin secretion both in vitro and in vivo by using a mathematical model. The model describes the triggering and amplifying pathways and reproduces a variety of in vitro and in vivo tests in subjects with and without T2D, identifies the mechanisms modulating first-phase insulin secretion rate in response to basal hyperglycemia or insulin resistance, and shows that β-cell dysfunction in T2D can be explained by an impaired amplifying pathway with no need to postulate defects in intracellular calcium handling.
Collapse
Affiliation(s)
- Eleonora Grespan
- Institute of Neuroscience, National Research Council, Padua, Italy
| | - Toni Giorgino
- Institute of Neuroscience, National Research Council, Padua, Italy
| | - Silva Arslanian
- Division of Weight Management, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA
- Division of Pediatric Endocrinology, Diabetes and Metabolism, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Andrea Natali
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Ele Ferrannini
- Institute of Clinical Physiology, National Research Council, Pisa, Italy
| | - Andrea Mari
- Institute of Neuroscience, National Research Council, Padua, Italy
| |
Collapse
|
29
|
Steven S, Hollingsworth KG, Small PK, Woodcock SA, Pucci A, Aribasala B, Al-Mrabeh A, Batterham RL, Taylor R. Calorie restriction and not glucagon-like peptide-1 explains the acute improvement in glucose control after gastric bypass in Type 2 diabetes. Diabet Med 2016; 33:1723-1731. [PMID: 27589584 DOI: 10.1111/dme.13257] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/31/2016] [Indexed: 12/15/2022]
Abstract
AIMS To compare directly the impact of glucagon-like peptide-1 secretion on glucose metabolism in individuals with Type 2 diabetes listed for Roux-en-Y gastric bypass surgery, randomized to be studied before and 7 days after undergoing Roux-en-Y gastric bypass or after following a very-low-calorie diet. METHODS A semi-solid meal test was used to investigate glucose, insulin and glucagon-like peptide-1 response. Insulin secretion in response to intravenous glucose and arginine stimulus was measured. Hepatic and pancreatic fat content was quantified using magnetic resonance imaging. RESULTS The decrease in fat mass was almost identical in the Roux-en-Y gastric bypass and the very-low-calorie diet groups (3.0±0.3 and 3.0±0.7kg). The early rise in plasma glucose level and in acute insulin secretion were greater after Roux-en-Y gastric bypass than after a very-low-calorie diet; however, the early rise in glucagon-like peptide-1 was disproportionately greater (sevenfold) after Roux-en-Y gastric bypass than after a very-low-calorie diet. This did not translate into a greater improvement in fasting glucose level or area under the curve for glucose. The reduction in liver fat was greater after Roux-en-Y gastric bypass (29.8±3.7 vs 18.6±4.0%) and the relationships between weight loss and reduction in liver fat differed between the Roux-en-Y gastric bypass group and the very-low-calorie diet group. CONCLUSIONS This study shows that gastroenterostomy increases the rate of nutrient absorption, bringing about a commensurately rapid rise in insulin level; however, there was no association with the large post-meal rise in glucagon-like peptide-1, and post-meal glucose homeostasis was similar in the Roux-en-Y gastric bypass and very-low-calorie diet groups. (Clinical trials registry number: ISRCTN11969319.).
Collapse
Affiliation(s)
- S Steven
- Magnetic Resonance Centre, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - K G Hollingsworth
- Magnetic Resonance Centre, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - P K Small
- Department of Surgery, Sunderland Royal Hospital, Sunderland, UK
| | - S A Woodcock
- Department of Surgery, North Tyneside General Hospital, North Shields, UK
| | - A Pucci
- Centre for Obesity Research, University College London, London, UK
| | - B Aribasala
- Computer Science Department, Faculty of Science, Lagos State University, Lagos, Nigeria
| | - A Al-Mrabeh
- Magnetic Resonance Centre, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - R L Batterham
- Centre for Obesity Research, University College London, London, UK
| | - R Taylor
- Magnetic Resonance Centre, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
30
|
Shankar SS, Vella A, Raymond RH, Staten MA, Calle RA, Bergman RN, Cao C, Chen D, Cobelli C, Dalla Man C, Deeg M, Dong JQ, Lee DS, Polidori D, Robertson RP, Ruetten H, Stefanovski D, Vassileva MT, Weir GC, Fryburg DA. Standardized Mixed-Meal Tolerance and Arginine Stimulation Tests Provide Reproducible and Complementary Measures of β-Cell Function: Results From the Foundation for the National Institutes of Health Biomarkers Consortium Investigative Series. Diabetes Care 2016; 39:1602-13. [PMID: 27407117 PMCID: PMC5001146 DOI: 10.2337/dc15-0931] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 06/15/2016] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Standardized, reproducible, and feasible quantification of β-cell function (BCF) is necessary for the evaluation of interventions to improve insulin secretion and important for comparison across studies. We therefore characterized the responses to, and reproducibility of, standardized methods of in vivo BCF across different glucose tolerance states. RESEARCH DESIGN AND METHODS Participants classified as having normal glucose tolerance (NGT; n = 23), prediabetes (PDM; n = 17), and type 2 diabetes mellitus (T2DM; n = 22) underwent two standardized mixed-meal tolerance tests (MMTT) and two standardized arginine stimulation tests (AST) in a test-retest paradigm and one frequently sampled intravenous glucose tolerance test (FSIGT). RESULTS From the MMTT, insulin secretion in T2DM was >86% lower compared with NGT or PDM (P < 0.001). Insulin sensitivity (Si) decreased from NGT to PDM (∼50%) to T2DM (93% lower [P < 0.001]). In the AST, insulin secretory response to arginine at basal glucose and during hyperglycemia was lower in T2DM compared with NGT and PDM (>58%; all P < 0.001). FSIGT showed decreases in both insulin secretion and Si across populations (P < 0.001), although Si did not differ significantly between PDM and T2DM populations. Reproducibility was generally good for the MMTT, with intraclass correlation coefficients (ICCs) ranging from ∼0.3 to ∼0.8 depending on population and variable. Reproducibility for the AST was very good, with ICC values >0.8 across all variables and populations. CONCLUSIONS Standardized MMTT and AST provide reproducible and complementary measures of BCF with characteristics favorable for longitudinal interventional trials use.
Collapse
Affiliation(s)
- Sudha S Shankar
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN
| | - Adrian Vella
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, MN
| | | | - Myrlene A Staten
- Kelly Government Solutions for National Institute of Diabetes and Digestive and Kidney Diseases, Rockville, MD
| | | | - Richard N Bergman
- Cedars-Sinai Diabetes and Obesity Research Institute, Los Angeles, CA
| | - Charlie Cao
- Takeda Development Center Americas, Deerfield, IL
| | | | - Claudio Cobelli
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Chiara Dalla Man
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Mark Deeg
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN
| | | | | | | | - R Paul Robertson
- Pacific Northwest Diabetes Research Institute, Seattle, WA Division of Endocrinology, Departments of Medicine and Pharmacology, University of Washington, Seattle, WA
| | | | | | | | | | | | | |
Collapse
|
31
|
Bizzotto R, Natali A, Gastaldelli A, Muscelli E, Krssak M, Brehm A, Roden M, Ferrannini E, Mari A. Glucose uptake saturation explains glucose kinetics profiles measured by different tests. Am J Physiol Endocrinol Metab 2016; 311:E346-57. [PMID: 27245333 DOI: 10.1152/ajpendo.00045.2016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 05/23/2016] [Indexed: 02/08/2023]
Abstract
It is known that for a given insulin level glucose clearance depends on glucose concentration. However, a quantitative representation of the concomitant effects of hyperinsulinemia and hyperglycemia on glucose clearance, necessary to describe heterogeneous tests such as euglycemic and hyperglycemic clamps and oral tests, is lacking. Data from five studies (123 subjects) using a glucose tracer and including all the above tests in normal and diabetic subjects were collected. A mathematical model was developed in which glucose utilization was represented as a Michaelis-Menten function of glucose with constant Km and insulin-controlled Vmax, consistently with the basic notions of glucose transport. Individual values for the model parameters were estimated using a population approach. Tracer data were accurately fitted in all tests. The estimated Km was 3.88 (2.83-5.32) mmol/l [median (interquartile range)]. Median model-derived glucose clearance at 600 pmol/l insulin was reduced from 246 to 158 ml·min(-1)·m(-2) when glucose was raised from 5 to 10 mmol/l. The model reproduced the characteristic lack of increase in glucose clearance when moderate hyperinsulinemia was accompanied by hyperglycemia. In all tests, insulin sensitivity was inversely correlated with BMI, as expected (R(2) = 0.234, P = 0.0001). In conclusion, glucose clearance in euglycemic and hyperglycemic clamps and oral tests can be described with a unifying model, consistent with the notions of glucose transport and able to reproduce the suppression of glucose clearance due to hyperglycemia observed in previous studies. The model may be important for the design of reliable glucose homeostasis simulators.
Collapse
Affiliation(s)
| | - Andrea Natali
- Department of Internal Medicine, University of Pisa School of Medicine, Pisa, Italy
| | | | - Elza Muscelli
- Department of Internal Medicine, University of Pisa School of Medicine, Pisa, Italy
| | | | | | - Michael Roden
- Heinrich-Heine University, Düsseldorf, Germany; German Diabetes Center, Düsseldorf, Germany; and German Center of Diabetes Research, München-Neuherberg, Germany
| | - Ele Ferrannini
- Department of Internal Medicine, University of Pisa School of Medicine, Pisa, Italy; CNR Institute of Clinical Physiology, Pisa, Italy
| | | |
Collapse
|
32
|
Seghieri M, Rebelos E, Astiarraga BD, Baldi S, Mari A, Ferrannini E. Impact of a mild decrease in fasting plasma glucose on β-cell function in healthy subjects and patients with type 2 diabetes. Am J Physiol Endocrinol Metab 2016; 310:E919-24. [PMID: 27072495 DOI: 10.1152/ajpendo.00428.2015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 04/04/2016] [Indexed: 01/09/2023]
Abstract
Restoring euglycaemia for weeks or months improves insulin secretion in patients with type 2 diabetes (T2D). We tested whether mild decrements in fasting glucose (FPG) acutely affect β-cell function and insulin sensitivity. Thirteen normotolerant (NGT) and 10 T2D patients volunteered in pairs. In an isoglycemic test (Iso), after 100 min of stabilization, an incremental glucose infusion over 3 h was applied to raise plasma glucose to >22 mmol/l, followed by an arginine challenge; in a subisoglycemic test (Sub), a glucose infusion matching the plasma glucose time course of Iso was preceded by an insulin infusion period (100 min) aimed at maintaining a mild FPG reduction while avoiding hypoglycaemia. β-Cell function was assessed by mathematical modeling, whereas the acute insulin response (AIR) to arginine was determined from C-peptide levels. In the Sub, FPG was lowered by 17% in NGT and 31% in T2D patients. On the glucose ramp, total insulin release was lower in Sub than in Iso in both groups [from 106 (43) to 75 (39) nmol/m(-2) in NGT and from 71 (63) to 64 (41) nmol/m(-2) in T2D, P = 0.001]. In the Sub, β-cell glucose sensitivity was significantly (P = 0.008) reduced in NGT [from 50 (31) to 43 (21) pmol·min(-1)·m(-2)·mM(-1)] but not in T2D [19 (20) to 20 (20) pmol·min(-1)·m(-2)·mM(-1)]. Likewise, AIR was lowered in NGT [8.9 (4.6) to 7.1 (4.4) nmol/l, P = 0.048] but not in T2D [4.7 (3.3) to 5.3 (3.2) nmol/l]. Insulin sensitivity improved in NGT but only marginally in T2D. Prestimulatory glucose levels acutely influence both β-cell function and insulin sensitivity differentially in nondiabetic and type 2 diabetic individuals.
Collapse
Affiliation(s)
- Marta Seghieri
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy;
| | - Eleni Rebelos
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Brenno D Astiarraga
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Simona Baldi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Andrea Mari
- National Research Council (CNR) Institute of Neuroscience,Padua, Italy; and
| | | |
Collapse
|
33
|
Steven S, Hollingsworth KG, Al-Mrabeh A, Avery L, Aribisala B, Caslake M, Taylor R. Very Low-Calorie Diet and 6 Months of Weight Stability in Type 2 Diabetes: Pathophysiological Changes in Responders and Nonresponders. Diabetes Care 2016; 39:808-15. [PMID: 27002059 DOI: 10.2337/dc15-1942] [Citation(s) in RCA: 249] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 01/27/2016] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Type 2 diabetes mellitus (T2DM) is generally regarded as an irreversible chronic condition. Because a very low-calorie diet (VLCD) can bring about acute return to normal glucose control in some people with T2DM, this study tested the potential durability of this normalization. The underlying mechanisms were defined. RESEARCH DESIGN AND METHODS People with a T2DM duration of 0.5-23 years (n = 30) followed a VLCD for 8 weeks. All oral agents or insulins were stopped at baseline. Following a stepped return to isocaloric diet, a structured, individualized program of weight maintenance was provided. Glucose control, insulin sensitivity, insulin secretion, and hepatic and pancreas fat content were quantified at baseline, after return to isocaloric diet, and after 6 months to permit the primary comparison of change between post-weight loss and 6 months in responders. Responders were defined as achieving fasting blood glucose <7 mmol/L after return to isocaloric diet. RESULTS Weight fell (98.0 ± 2.6 to 83.8 ± 2.4 kg) and remained stable over 6 months (84.7 ± 2.5 kg). Twelve of 30 participants achieved fasting plasma glucose <7 mmol/L after return to isocaloric diet (responders), and 13 of 30 after 6 months. Responders had a shorter duration of diabetes and a higher initial fasting plasma insulin level. HbA1c fell from 7.1 ± 0.3 to 5.8 ± 0.2% (55 ± 4 to 40 ± 2 mmol/mol) in responders (P < 0.001) and from 8.4 ± 0.3 to 8.0 ± 0.5% (68 ± 3 to 64 ± 5 mmol/mol) in nonresponders, remaining constant at 6 months (5.9 ± 0.2 and 7.8 ± 0.3% [41 ± 2 and 62 ± 3 mmol/mol], respectively). The responders were characterized by return of first-phase insulin response. CONCLUSIONS A robust and sustainable weight loss program achieved continuing remission of diabetes for at least 6 months in the 40% who responded to a VLCD by achieving fasting plasma glucose of <7 mmol/L. T2DM is a potentially reversible condition.
Collapse
Affiliation(s)
- Sarah Steven
- Magnetic Resonance Centre, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K
| | - Kieren G Hollingsworth
- Magnetic Resonance Centre, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K
| | - Ahmad Al-Mrabeh
- Magnetic Resonance Centre, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K
| | - Leah Avery
- MoveLab, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K
| | - Benjamin Aribisala
- Computer Science Department, Faculty of Science, Lagos State University, Lagos, Nigeria
| | - Muriel Caslake
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, U.K
| | - Roy Taylor
- Magnetic Resonance Centre, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K.
| |
Collapse
|
34
|
Steven S, Hollingsworth KG, Small PK, Woodcock SA, Pucci A, Aribisala B, Al-Mrabeh A, Daly AK, Batterham RL, Taylor R. Weight Loss Decreases Excess Pancreatic Triacylglycerol Specifically in Type 2 Diabetes. Diabetes Care 2016; 39:158-65. [PMID: 26628414 DOI: 10.2337/dc15-0750] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 07/22/2015] [Indexed: 02/03/2023]
Abstract
OBJECTIVE This study determined whether the decrease in pancreatic triacylglycerol during weight loss in type 2 diabetes mellitus (T2DM) is simply reflective of whole-body fat or specific to diabetes and associated with the simultaneous recovery of insulin secretory function. RESEARCH DESIGN AND METHODS Individuals listed for gastric bypass surgery who had T2DM or normal glucose tolerance (NGT) matched for age, weight, and sex were studied before and 8 weeks after surgery. Pancreas and liver triacylglycerol were quantified using in-phase, out-of-phase MRI. Also measured were the first-phase insulin response to a stepped intravenous glucose infusion, hepatic insulin sensitivity, and glycemic and incretin responses to a semisolid test meal. RESULTS Weight loss after surgery was similar (NGT: 12.8 ± 0.8% and T2DM: 13.6 ± 0.7%) as was the change in fat mass (56.7 ± 3.3 to 45.4 ± 2.3 vs. 56.6 ± 2.4 to 43.0 ± 2.4 kg). Pancreatic triacylglycerol did not change in NGT (5.1 ± 0.2 to 5.5 ± 0.4%) but decreased in the group with T2DM (6.6 ± 0.5 to 5.4 ± 0.4%; P = 0.007). First-phase insulin response to a stepped intravenous glucose infusion did not change in NGT (0.24 [0.13-0.46] to 0.23 [0.19-0.37] nmol ⋅ min(-1) ⋅ m(-2)) but normalized in T2DM (0.08 [-0.01 to -0.10] to 0.22 [0.07-0.30]) nmol ⋅ min(-1) ⋅ m(-2) at week 8 (P = 0.005). No differential effect of incretin secretion was observed after gastric bypass, with more rapid glucose absorption bringing about equivalently enhanced glucagon-like peptide 1 secretion in the two groups. CONCLUSIONS The fall in intrapancreatic triacylglycerol in T2DM, which occurs during weight loss, is associated with the condition itself rather than decreased total body fat.
Collapse
Affiliation(s)
- Sarah Steven
- Magnetic Resonance Centre, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K
| | - Kieren G Hollingsworth
- Magnetic Resonance Centre, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K. Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K
| | - Peter K Small
- Department of Surgery, Sunderland Royal Hospital, Sunderland, U.K
| | - Sean A Woodcock
- Department of Surgery, North Tyneside General Hospital, North Shields, U.K
| | - Andrea Pucci
- Centre for Obesity Research, University College London, London, U.K
| | - Benjamin Aribisala
- Computer Science Department, Faculty of Science, Lagos State University, Lagos, Nigeria
| | - Ahmad Al-Mrabeh
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K
| | - Ann K Daly
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K
| | | | - Roy Taylor
- Magnetic Resonance Centre, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K. Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K.
| |
Collapse
|
35
|
Acute intravenous glucose load impairs early insulin secretion and insulin content in islet β cells in mice. Life Sci 2016; 144:148-55. [DOI: 10.1016/j.lfs.2015.12.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 11/14/2015] [Accepted: 12/04/2015] [Indexed: 01/04/2023]
|
36
|
De Gaetano A, Gaz C, Palumbo P, Panunzi S. A Unifying Organ Model of Pancreatic Insulin Secretion. PLoS One 2015; 10:e0142344. [PMID: 26555895 PMCID: PMC4640662 DOI: 10.1371/journal.pone.0142344] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 10/20/2015] [Indexed: 12/25/2022] Open
Abstract
The secretion of insulin by the pancreas has been the object of much attention over the past several decades. Insulin is known to be secreted by pancreatic β-cells in response to hyperglycemia: its blood concentrations however exhibit both high-frequency (period approx. 10 minutes) and low-frequency oscillations (period approx. 1.5 hours). Furthermore, characteristic insulin secretory response to challenge maneuvers have been described, such as frequency entrainment upon sinusoidal glycemic stimulation; substantial insulin peaks following minimal glucose administration; progressively strengthened insulin secretion response after repeated administration of the same amount of glucose; insulin and glucose characteristic curves after Intra-Venous administration of glucose boli in healthy and pre-diabetic subjects as well as in Type 2 Diabetes Mellitus. Previous modeling of β-cell physiology has been mainly directed to the intracellular chain of events giving rise to single-cell or cell-cluster hormone release oscillations, but the large size, long period and complex morphology of the diverse responses to whole-body glucose stimuli has not yet been coherently explained. Starting with the seminal work of Grodsky it was hypothesized that the population of pancreatic β-cells, possibly functionally aggregated in islets of Langerhans, could be viewed as a set of independent, similar, but not identical controllers (firing units) with distributed functional parameters. The present work shows how a single model based on a population of independent islet controllers can reproduce very closely a diverse array of actually observed experimental results, with the same set of working parameters. The model's success in reproducing a diverse array of experiments implies that, in order to understand the macroscopic behaviour of the endocrine pancreas in regulating glycemia, there is no need to hypothesize intrapancreatic pacemakers, influences between different islets of Langerhans, glycolitic-induced oscillations or β-cell sensitivity to the rate of change of glycemia.
Collapse
Affiliation(s)
- Andrea De Gaetano
- CNR-IASI BioMatLab (Italian National Research Council - Institute of Analysis, Systems and Computer Science - Biomathematics Laboratory), UCSC Largo A. Gemelli 8, 00168 Rome, Italy
| | - Claudio Gaz
- CNR-IASI BioMatLab (Italian National Research Council - Institute of Analysis, Systems and Computer Science - Biomathematics Laboratory), UCSC Largo A. Gemelli 8, 00168 Rome, Italy
- Sapienza Università di Roma, Department of Computer, Control and Management Engineering (DIAG), Via Ariosto 25, 00185 Rome, Italy
| | - Pasquale Palumbo
- CNR-IASI BioMatLab (Italian National Research Council - Institute of Analysis, Systems and Computer Science - Biomathematics Laboratory), UCSC Largo A. Gemelli 8, 00168 Rome, Italy
| | - Simona Panunzi
- CNR-IASI BioMatLab (Italian National Research Council - Institute of Analysis, Systems and Computer Science - Biomathematics Laboratory), UCSC Largo A. Gemelli 8, 00168 Rome, Italy
| |
Collapse
|
37
|
Henquin JC, Dufrane D, Kerr-Conte J, Nenquin M. Dynamics of glucose-induced insulin secretion in normal human islets. Am J Physiol Endocrinol Metab 2015; 309:E640-50. [PMID: 26264556 DOI: 10.1152/ajpendo.00251.2015] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 08/10/2015] [Indexed: 11/22/2022]
Abstract
The biphasic pattern of glucose-induced insulin secretion is altered in type 2 diabetes. Impairment of the first phase is an early sign of β-cell dysfunction, but the underlying mechanisms are still unknown. Their identification through in vitro comparisons of islets from diabetic and control subjects requires characterization and quantification of the dynamics of insulin secretion by normal islets. When perifused normal human islets were stimulated with 15 mmol/l glucose (G15), the proinsulin/insulin ratio in secretory products rapidly and reversibly decreased (∼50%) and did not reaugment with time. Switching from prestimulatory G3 to G6-G30 induced biphasic insulin secretion with flat but sustained (2 h) second phases. Stimulation index reached 6.7- and 3.6-fold for the first and second phases induced by G10. Concentration dependency was similar for both phases, with half-maximal and maximal responses at G6.5 and G15, respectively. First-phase response to G15-G30 was diminished by short (30-60 min) prestimulation in G6 (vs. G3) and abolished by prestimulation in G8, whereas the second phase was unaffected. After 1-2 days of culture in G8 (instead of G5), islets were virtually unresponsive to G15. In both settings, a brief return to G3-G5 or transient omission of CaCl2 restored biphasic insulin secretion. Strikingly, tolbutamide and arginine evoked immediate insulin secretion in islets refractory to glucose. In conclusion, we quantitatively characterized the dynamics of glucose-induced insulin secretion in normal human islets and showed that slight elevation of prestimulatory glucose reversibly impairs the first phase, which supports the view that the similar impairment in type 2 diabetic patients might partially be a secondary phenomenon.
Collapse
Affiliation(s)
- Jean-Claude Henquin
- Unit of Endocrinology and Metabolism, Faculty of Medicine, University of Louvain, Brussels, Belgium;
| | - Denis Dufrane
- Endocrine Cell Therapy Unit, University Clinics Saint-Luc, University of Louvain, Brussels, Belgium
| | - Julie Kerr-Conte
- Institut National de la Santé et de la Recherche Médicale U1190, Translational Research for Diabetes, and European Genomic Institute for Diabetes, University of Lille, Lille, France
| | - Myriam Nenquin
- Unit of Endocrinology and Metabolism, Faculty of Medicine, University of Louvain, Brussels, Belgium
| |
Collapse
|
38
|
Qinna NA, Badwan AA. Impact of streptozotocin on altering normal glucose homeostasis during insulin testing in diabetic rats compared to normoglycemic rats. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:2515-25. [PMID: 26005328 PMCID: PMC4427609 DOI: 10.2147/dddt.s79885] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Streptozotocin (STZ) is currently the most used diabetogenic agent in testing insulin and new antidiabetic drugs in animals. Due to the toxic and disruptive nature of STZ on organs, apart from pancreas, involved in preserving the body’s normal glucose homeostasis, this study aims to reassess the action of STZ in inducing different glucose response states in diabetic rats while testing insulin. Diabetic Sprague-Dawley rats induced with STZ were classified according to their initial blood glucose levels into stages. The effect of randomizing rats in such a manner was investigated for the severity of interrupting normal liver, pancreas, and kidney functions. Pharmacokinetic and pharmacodynamic actions of subcutaneously injected insulin in diabetic and nondiabetic rats were compared. Interruption of glucose homeostasis by STZ was challenged by single and repeated administrations of injected insulin and oral glucose to diabetic rats. In diabetic rats with high glucose (451–750 mg/dL), noticeable changes were seen in the liver and kidney functions compared to rats with lower basal glucose levels. Increased serum levels of recombinant human insulin were clearly indicated by a significant increase in the calculated maximum serum concentration and area under the concentration–time curve. Reversion of serum glucose levels to normal levels pre- and postinsulin and oral glucose administrations to STZ diabetic rats were found to be variable. In conclusion, diabetic animals were more responsive to insulin than nondiabetic animals. STZ was capable of inducing different levels of normal glucose homeostasis disruption in rats. Both pharmacokinetic and pharmacodynamic actions of insulin were altered when different initial blood glucose levels of STZ diabetic rats were selected for testing. Such findings emphasize the importance of selecting predefined and unified glucose levels when using STZ as a diabetogenic agent in experimental protocols evaluating new antidiabetic agents and insulin delivery systems.
Collapse
Affiliation(s)
- Nidal A Qinna
- Department of Pharmacology and Biomedical Sciences, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, Jordan
| | - Adnan A Badwan
- Research and Innovation Centre, The Jordanian Pharmaceutical Manufacturing Co. Plc. (JPM), Amman, Jordan
| |
Collapse
|
39
|
Merovci A, Mari A, Solis-Herrera C, Xiong J, Daniele G, Chavez-Velazquez A, Tripathy D, Urban McCarthy S, Abdul-Ghani M, DeFronzo RA. Dapagliflozin lowers plasma glucose concentration and improves β-cell function. J Clin Endocrinol Metab 2015; 100:1927-32. [PMID: 25710563 PMCID: PMC4422889 DOI: 10.1210/jc.2014-3472] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND β-Cell dysfunction is a core defect in T2DM, and chronic, sustained hyperglycemia has been implicated in progressive β-cell failure, ie, glucotoxicity. The aim of the present study was to examine the effect of lowering the plasma glucose concentration with dapagliflozin, a glucosuric agent, on β-cell function in T2DM individuals. RESEARCH DESIGN AND METHODS Twenty-four subjects with T2DM received dapagliflozin (n = 16) or placebo (n = 8) for 2 weeks, and a 75-g oral glucose tolerance test (OGTT) and insulin clamp were performed before and after treatment. Plasma glucose, insulin, and C-peptide concentrations were measured during the OGTT. RESULTS Dapagliflozin significantly lowered both the fasting and 2-hour plasma glucose concentrations and the incremental area under the plasma glucose concentration curve (ΔG0-120) during OGTT by -33 ± 5 mg/dL, -73 ± 9 mg/dL, and -60 ± 12 mg/dL · min, respectively, compared to -13 ± 9, -33 ± 13, and -18 ± 9 reductions in placebo-treated subjects (both P < .01). The incremental area under the plasma C-peptide concentration curve tended to increase in dapagliflozin-treated subjects, whereas it did not change in placebo-treated subjects. Thus, ΔC-Pep0-120/ΔG0-120 increased significantly in dapagliflozin-treated subjects, whereas it did not change in placebo-treated subjects (0.019 ± 0.005 vs 0.002 ± 0.006; P < .01). Dapagliflozin significantly improved whole-body insulin sensitivity (insulin clamp). Thus, β-cell function, measured as ΔC-Pep0-120/ ΔG0-120 ÷ insulin resistance, increased by 2-fold (P < .01) in dapagliflozin-treated vs placebo-treated subjects. CONCLUSION Lowering the plasma glucose concentration with dapagliflozin markedly improves β-cell function, providing strong support in man for the glucotoxic effect of hyperglycemia on β-cell function.
Collapse
Affiliation(s)
- Aurora Merovci
- Division of Diabetes (A.Me., C.S., G.D., A.C.-V., D.T., S.U.M., M.A.-G., R.A.D.), University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229; and Consiglio Nazionale delle Ricerche Institute of Biomedical Engineering (A.Ma.), 35127 Padova, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Kolka CM, Castro AVB, Kirkman EL, Bergman RN. Modest hyperglycemia prevents interstitial dispersion of insulin in skeletal muscle. Metabolism 2015; 64:330-7. [PMID: 25468139 PMCID: PMC4277905 DOI: 10.1016/j.metabol.2014.10.036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 09/22/2014] [Accepted: 10/28/2014] [Indexed: 10/24/2022]
Abstract
UNLABELLED Insulin injected directly into skeletal muscle diffuses rapidly through the interstitial space to cause glucose uptake, but this is blocked in insulin resistance. As glucotoxicity is associated with endothelial dysfunction, the observed hyperglycemia in diet-induced obese dogs may inhibit insulin access to muscle cells, and exacerbate insulin resistance. Here we asked whether interstitial insulin diffusion is reduced in modest hyperglycemia, similar to that induced by a high fat diet. METHODS During normoglycemic (100 mg/dl) and moderately hyperglycemic (120 mg/dl) clamps in anesthetized canines, sequential doses of insulin were injected into the vastus medialis of one hindlimb; the contra-lateral limb served as a control. Plasma samples were collected and analyzed for insulin content. Lymph vessels of the hind leg were also catheterized, and lymph samples were analyzed as an indicator of interstitial insulin concentration. RESULTS Insulin injection increased lymph insulin in normoglycemic animals, but not in hyperglycemic animals. Muscle glucose uptake was elevated in response to hyperglycemia, however the insulin-mediated glucose uptake in normoglycemic controls was not observed in hyperglycemia. Modest hyperglycemia prevented intra-muscularly injected insulin from diffusing through the interstitial space reduced insulin-mediated glucose uptake. CONCLUSION Hyperglycemia prevents the appearance of injected insulin in the interstitial space, thus reducing insulin action on skeletal muscle cells.
Collapse
MESH Headings
- Absorption, Physiological
- Animals
- Biological Transport/drug effects
- Diffusion
- Dogs
- Dose-Response Relationship, Drug
- Extracellular Space/chemistry
- Glucose/metabolism
- Glucose Clamp Technique
- Hindlimb
- Hyperglycemia/blood
- Hyperglycemia/drug therapy
- Hyperglycemia/metabolism
- Hyperglycemia/physiopathology
- Hypoglycemic Agents/administration & dosage
- Hypoglycemic Agents/metabolism
- Hypoglycemic Agents/pharmacokinetics
- Hypoglycemic Agents/therapeutic use
- Injections, Intramuscular
- Insulin Resistance
- Insulin, Regular, Pork/administration & dosage
- Insulin, Regular, Pork/analysis
- Insulin, Regular, Pork/pharmacokinetics
- Insulin, Regular, Pork/therapeutic use
- Lymph/chemistry
- Lymph/drug effects
- Male
- Quadriceps Muscle/chemistry
- Quadriceps Muscle/drug effects
- Quadriceps Muscle/metabolism
- Severity of Illness Index
- Tissue Distribution
Collapse
Affiliation(s)
- Cathryn M Kolka
- Department of Physiology and Biophysics, University of Southern California, Los Angeles, CA.
| | - Ana Valeria B Castro
- Department of Physiology and Biophysics, University of Southern California, Los Angeles, CA
| | - Erlinda L Kirkman
- Department of Physiology and Biophysics, University of Southern California, Los Angeles, CA
| | - Richard N Bergman
- Department of Physiology and Biophysics, University of Southern California, Los Angeles, CA
| |
Collapse
|
41
|
Declining ß-cell function is associated with the lack of long-range negative correlation in glucose dynamics and increased glycemic variability: A retrospective analysis in patients with type 2 diabetes. JOURNAL OF CLINICAL AND TRANSLATIONAL ENDOCRINOLOGY 2014; 1:192-199. [PMID: 29159101 PMCID: PMC5685022 DOI: 10.1016/j.jcte.2014.09.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 09/01/2014] [Accepted: 09/12/2014] [Indexed: 11/24/2022]
Abstract
Objective To determine whether characteristics of glucose dynamics are reflections of β-cell function or rather of inadequate diabetes control. Materials/methods We analyzed historical liquid meal tolerance test (LMTT) and continuous glucose monitoring (CGM) data, which had been obtained from 56 non-insulin treated type 2 diabetic outpatients during withdrawal of antidiabetic drugs. Computed CGM parameters included detrended fluctuation analysis (DFA)-based indices, autocorrelation function exponent, mean amplitude of glycemic excursions (MAGE), glucose SD, and measures of glycemic exposure. The LMTT-based disposition index (LMTT-DI) calculated from the ratio of the area-under-the-insulin-curve to the area-under-the-glucose-curve and Matsuda index was used to assess relationships among β-cell function, glucose profile complexity, autocorrelation function, and glycemic variability. Results The LMTT-DI was inverse linearly correlated with the short-range α1 and long-range scaling exponent α2 (r = −0.275 and −0.441, respectively, p < 0.01) such that lower glucose complexity was associated with better preserved insulin reserve, but it did not correlate with the autocorrelation decay exponent γ. By contrast, the LMTT-DI was strongly correlated with MAGE and SD (r = 0.625 and 0.646, both p < 0.001), demonstrating a curvilinear relationship between β-cell function and glycemic variability. On stepwise regression analyses, the LMTT-DI emerged as an independent contributor, explaining 20, 38, and 47% (all p < 0.001) of the variance in the long-range DFA scaling exponent, MAGE, and hemoglobin A1C, respectively, whereas insulin sensitivity failed to contribute independently. Conclusions Loss of complexity and increased variability in glucose profiles are associated with declining β-cell reserve and worsening glycemic control.
Collapse
Key Words
- ACF, autocorrelation function
- AUC, area under the curve
- CGM, continuous glucose monitoring
- Cp, C-peptide
- DFA, detrended fluctuation analysis
- Disposition index
- Glucose profile dynamics
- LMTT, liquid meal tolerance test
- LMTT-DI, LMTT-based disposition index
- MAGE, mean amplitude of glycemic excursions
- OHA, oral hypoglycemic agent
- SD, standard deviation
- TZDs, thiazolidinediones
- Type 2 diabetes
- β-Cell reserve
Collapse
|
42
|
Ferrannini E, Mari A. β-Cell function in type 2 diabetes. Metabolism 2014; 63:1217-27. [PMID: 25070616 DOI: 10.1016/j.metabol.2014.05.012] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 05/09/2014] [Accepted: 05/25/2014] [Indexed: 01/09/2023]
Abstract
Different in vivo tests explore different aspects of β-cell function. Because intercorrelation of insulin secretion indices is modest, no single in vivo test allows β-cell function to be assessed with accuracy and specificity comparable to insulin sensitivity. Physiologically-based mathematical modeling is necessary to interpret insulin secretory responses in terms of relevant parameters of β-cell function. Models can be used to analyze intravenous glucose tests, but secretory responses to intravenous glucose may be paradoxical in subjects with diabetes. Use of oral glucose (or mixed meal) data may be preferable not only for simplicity but also for physiological interpretation. While the disposition index focuses on the relationship between insulin secretion and insulin resistance, secretion parameters reflecting the dynamic response to changing glucose levels over a time frame of minutes or hours--such as β-cell glucose sensitivity--are key to explain changes in glucose tolerance and are largely independent of insulin sensitivity. Pathognomonic of the β-cell defect of type 2 diabetes is a reduced glucose sensitivity, which is accompanied by normal or raised absolute insulin secretion rates--compensatory to the attendant insulin resistance--and impaired incretin-induced potentiation. As β-cell mass is frequently within the range of nondiabetic individuals, these defects are predominantly functional and potentially reversible. Any intervention, on lifestyle or with drugs, that improves glucose tolerance does so primarily through increased β-cell glucose sensitivity. So far, however, no intervention has proven unequivocally capable of modifying the natural course of β-cell dysfunction.
Collapse
Affiliation(s)
- Ele Ferrannini
- Department of Clinical & Experimental Medicine, University of Pisa, Italy.
| | - Andrea Mari
- C N R Institute of Biomedical Engineering, Padova, Italy
| |
Collapse
|
43
|
Docherty PD, Chase JG, Te Morenga L, Fisk LM. A novel hierarchal-based approach to measure insulin sensitivity and secretion in at-risk populations. J Diabetes Sci Technol 2014; 8:807-14. [PMID: 24876451 PMCID: PMC4764222 DOI: 10.1177/1932296814536511] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The pathogenesis of type 2 diabetes is characterized by insulin resistance and insulin secretory dysfunction. Few existing metabolic tests measure both characteristics, and no such tests are inexpensive enough to enable widespread use. A hierarchical approach uses 2 down-sampled tests in the dynamic insulin sensitivity and secretion test (DISST) family to first determine insulin sensitivity (SI) using 4 glucose measurements. Second the insulin secretion is determined for only participants with reduced SI using 3 C-peptide measurements from the original test. The hierarchical approach is assessed via its ability to classify 214 individual test responses of 71 females with an elevated risk of type 2 diabetes into 5 bins with equivalence to the fully sampled DISST. Using an arbitrary SI cut-off, 102 test responses were reassayed for C-peptide and unique insulin secretion characteristics estimated. The hierarchical approach correctly classified 84.5% of the test responses and 94.4% of the responses of individuals with increased fasting glucose. The hierarchical approach is a low-cost methodology for measuring key characteristics of type 2 diabetes. Thus the approach could provide an economical approach to studying the pathogenesis of type 2 diabetes, or in early risk screening. As the higher cost test uses the same clinical protocol as the low-cost test, the cost of the additional information is limited to the assay cost of C-peptide, and no additional procedures or callbacks are required.
Collapse
|
44
|
Baack ML, Wang C, Hu S, Segar JL, Norris AW. Hyperglycemia induces embryopathy, even in the absence of systemic maternal diabetes: an in vivo test of the fuel mediated teratogenesis hypothesis. Reprod Toxicol 2014; 46:129-36. [PMID: 24721120 DOI: 10.1016/j.reprotox.2014.03.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 03/31/2014] [Accepted: 03/31/2014] [Indexed: 12/29/2022]
Abstract
Embryonic exposure to excess circulating fuels is proposed to underlie diabetic embryopathy. To isolate the effects of hyperglycemia from the many systemic anomalies of diabetes, we infused 4 mg/min glucose into the left uterine artery of non-diabetic pregnant rats on gestation days (GD) 7-9. Right-sided embryos and dams exhibited no glucose elevation. Embryos were assessed on GD13, comparing the left versus right uterine horns. Hyperglycemic exposure increased rates of embryopathy, resorptions, and worsened embryopathy severity. By contrast, saline infusion did not affect any of these parameters. To assess for possible embryopathy susceptibility bias between uterine horns, separate dams were given retinoic acid (25mg/kg, a mildly embryopathic dose) systemically on GD7.5. The resultant embryopathy rates were equivalent between uterine horns. We conclude that hyperglycemia, even in the absence of systemic maternal diabetes, is sufficient to produce in vivo embryopathy during organogenesis.
Collapse
Affiliation(s)
- Michelle L Baack
- University of Iowa Children's Hospital, Department of Pediatrics, Iowa City, IA, USA
| | - Chunlin Wang
- University of Iowa Children's Hospital, Department of Pediatrics, Iowa City, IA, USA
| | - Shanming Hu
- University of Iowa Children's Hospital, Department of Pediatrics, Iowa City, IA, USA
| | - Jeffrey L Segar
- University of Iowa Children's Hospital, Department of Pediatrics, Iowa City, IA, USA
| | - Andrew W Norris
- University of Iowa Children's Hospital, Department of Pediatrics, Iowa City, IA, USA.
| |
Collapse
|
45
|
Meier JJ, Bonadonna RC. Role of reduced β-cell mass versus impaired β-cell function in the pathogenesis of type 2 diabetes. Diabetes Care 2013; 36 Suppl 2:S113-9. [PMID: 23882035 PMCID: PMC3920783 DOI: 10.2337/dcs13-2008] [Citation(s) in RCA: 170] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Juris J Meier
- Division of Diabetology and Gastrointestinal Endocrinology, St. Josef-Hospital, Ruhr-University of Bochum, Bochum, Germany.
| | | |
Collapse
|
46
|
Farrow HA, Rand JS, Morton JM, O'Leary CA, Sunvold GD. Effect of dietary carbohydrate, fat, and protein on postprandial glycemia and energy intake in cats. J Vet Intern Med 2013; 27:1121-35. [PMID: 23869495 DOI: 10.1111/jvim.12139] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 03/13/2013] [Accepted: 05/29/2013] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Reducing carbohydrate intake is recommended in diabetic cats and might also be useful in some healthy cats to decrease diabetes risk. OBJECTIVE To compare postprandial glucose and insulin concentrations and energy intakes between cats fed diets high in protein, fat, or carbohydrate. ANIMALS Twenty-four lean cats with normal glucose tolerance. METHODS In a prospective randomized study, each of 3 matched groups (n = 8) received a different test diet for 5 weeks. Diets were high in either protein (46% of metabolizable energy [ME]), fat (47% ME), or carbohydrate (47% ME). Glucose and insulin were measured during glucose tolerance, ad libitum, and meal-feeding tests. RESULTS During ad libitum feeding, cats fed the high-carbohydrate diet consumed 25% and 18% more carbohydrate than cats fed diets high in fat and protein, respectively, and energy intake was highest when the high-fat and high-protein diets were fed. Regardless of the feeding pattern, cats fed the high-carbohydrate diet had 10-31% higher peak and mean glucose compared with both other diets; peak glucose in some cats reached 10.4 mmol/L (188 mg/dL) in cats fed 47% ME carbohydrate and 9.0 mmol/L (162 mg/dL) in cats fed 23% ME. CONCLUSIONS AND CLINICAL IMPORTANCE High-carbohydrate diets increase postprandial glycemia in healthy cats compared with diets high in fat or protein, although energy intake is lower. Avoidance of high- and moderate-carbohydrate diets can be advantageous in cats at risk of diabetes. Maintenance energy requirements should be fed to prevent weight gain when switching to lower carbohydrate diets.
Collapse
Affiliation(s)
- H A Farrow
- School of Veterinary Science, The University of Queensland, Brisbane, Qld, Australia
| | | | | | | | | |
Collapse
|
47
|
Trombetta M, Boselli L, Cretti A, Calì A, Vettore M, Caruso B, Dorizzi R, Avogaro A, Muggeo M, Bonora E, Bonadonna RC. Type 2 diabetes mellitus: a disease of the governance of the glucose-insulin system: an experimental metabolic control analysis study. Nutr Metab Cardiovasc Dis 2013; 23:23-30. [PMID: 21937205 DOI: 10.1016/j.numecd.2011.05.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Revised: 04/28/2011] [Accepted: 05/06/2011] [Indexed: 11/21/2022]
Abstract
BACKGROUND AND AIMS The relatives role of each component of the glucose-insulin system in determining hyperglycemia in type 2 diabetes is still under debate. Metabolic Control Analysis (MCA) quantifies the control exerted by each component of a system on a variable of interest, by computing the relevant coefficients of control (CCs), which are systemic properties. We applied MCA to the intravenous glucose tolerance test (IVGTT) to quantify the CCs of the main components of the glucose-insulin system on intravenous glucose tolerance. METHODS AND RESULTS We combined in vivo phenotyping (IVGTT/euglycaemic insulin clamp) and in silico modeling (GLUKINSLOOP.1) to compute the CCs of intravenous glucose tolerance in healthy insulin-sensitive (n = 9, NGR-IS), healthy insulin-resistant (n = 7, NGR-IR) and subdiabetic hyperglycemic (n = 8, PreT2DM) individuals and in patients with newly diagnosed type 2 diabetes (n = 7, T2DM). Altered insulin secretion and action were documented in NGR-IR and PreT2DM groups, but only 1st phase insulin secretion was significantly lower in T2DM than in PreT2DM (p < 0.05). The CCs changed little in the nondiabetic groups. However, several CCs were significantly altered in the patients (e.g. CCs of beta cell: -0.75 ± 0.10, -0.64 ± 0.15, -0.56 ± 0.09 and -0.19 ± 0.04 in NGR-IS, NGR-IR, PreT2DM and T2DM, respectively; p < 0.01 by MANOVA), and they could not be corrected by matching in silico nondiabetic and T2DM groups for 1st phase secretion. CONCLUSIONS Type 2 diabetes is characterized not only by loss of function of the elements of the glucose-insulin system, but also by changes in systemic properties (CCs). As such, it could be considered a disease of the governance of the glucose-insulin system.
Collapse
Affiliation(s)
- M Trombetta
- Department of Medicine, Section of Endocrinology, University of Verona School of Medicine, Verona, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Verkest KR, Rand JS, Fleeman LM, Morton JM. Spontaneously obese dogs exhibit greater postprandial glucose, triglyceride, and insulin concentrations than lean dogs. Domest Anim Endocrinol 2012; 42:103-12. [PMID: 22130330 DOI: 10.1016/j.domaniend.2011.10.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Revised: 10/04/2011] [Accepted: 10/08/2011] [Indexed: 10/15/2022]
Abstract
Dogs do not appear to progress from obesity-induced insulin resistance to type 2 diabetes mellitus. Both postprandial hyperglycemia and postprandial hypertriglyceridemia have been proposed to cause or maintain beta cell failure and progression to type 2 diabetes mellitus in other species. Postprandial glucose, triglyceride, and insulin concentrations have not been compared in lean and obese dogs. We measured serum glucose, triglyceride, and insulin concentrations in nine naturally occurring obese and nine age- and gender-matched lean dogs. After a 24-h fast, dogs were fed half their calculated daily energy requirement of a standardized diet that provided 37% and 40% of metabolizable energy as carbohydrate and fat, respectively. Fasting and postprandial glucose and triglyceride concentrations were greater in the obese dogs (P < 0.001), although the mean insulin concentration for this group was five times greater than that of the lean group (P < 0.001). Most of the 0.6 mM (11 mg/dL) difference in mean postprandial glucose concentrations between lean and obese dogs was attributable to a subset of persistently hyperglycemic obese dogs with mean postprandial glucose concentrations 1.0 mM (18 mg/dL) greater than that in lean dogs. Persistently hyperglycemic obese dogs had lower triglyceride (P = 0.02 to 0.04) and insulin (P < 0.02) concentrations than other obese dogs. None of the dogs developed clinical signs of diabetes mellitus during follow-up for a median of 2.6 yr. We conclude that pancreatic beta cells in dogs are either not sensitive to toxicity because of mild hyperglycemia or lack another component of the pathophysiology of beta cell failure in type 2 diabetes mellitus.
Collapse
Affiliation(s)
- K R Verkest
- Centre for Companion Animal Health, School of Veterinary Science, The University of Queensland, Queensland, Australia.
| | | | | | | |
Collapse
|
49
|
Lim EL, Hollingsworth KG, Aribisala BS, Chen MJ, Mathers JC, Taylor R. Reversal of type 2 diabetes: normalisation of beta cell function in association with decreased pancreas and liver triacylglycerol. Diabetologia 2011; 54:2506-14. [PMID: 21656330 PMCID: PMC3168743 DOI: 10.1007/s00125-011-2204-7] [Citation(s) in RCA: 731] [Impact Index Per Article: 56.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Accepted: 05/05/2011] [Indexed: 12/19/2022]
Abstract
AIMS/HYPOTHESIS Type 2 diabetes is regarded as inevitably progressive, with irreversible beta cell failure. The hypothesis was tested that both beta cell failure and insulin resistance can be reversed by dietary restriction of energy intake. METHODS Eleven people with type 2 diabetes (49.5 ± 2.5 years, BMI 33.6 ± 1.2 kg/m(2), nine male and two female) were studied before and after 1, 4 and 8 weeks of a 2.5 MJ (600 kcal)/day diet. Basal hepatic glucose output, hepatic and peripheral insulin sensitivity and beta cell function were measured. Pancreas and liver triacylglycerol content was measured using three-point Dixon magnetic resonance imaging. An age-, sex- and weight-matched group of eight non-diabetic participants was studied. RESULTS After 1 week of restricted energy intake, fasting plasma glucose normalised in the diabetic group (from 9.2 ± 0.4 to 5.9 ± 0.4 mmol/l; p = 0.003). Insulin suppression of hepatic glucose output improved from 43 ± 4% to 74 ± 5% (p = 0.003 vs baseline; controls 68 ± 5%). Hepatic triacylglycerol content fell from 12.8 ± 2.4% in the diabetic group to 2.9 ± 0.2% by week 8 (p = 0.003). The first-phase insulin response increased during the study period (0.19 ± 0.02 to 0.46 ± 0.07 nmol min(-1) m(-2); p < 0.001) and approached control values (0.62 ± 0.15 nmol min(-1) m(-2); p = 0.42). Maximal insulin response became supranormal at 8 weeks (1.37 ± 0.27 vs controls 1.15 ± 0.18 nmol min(-1) m(-2)). Pancreatic triacylglycerol decreased from 8.0 ± 1.6% to 6.2 ± 1.1% (p = 0.03). CONCLUSIONS/INTERPRETATION Normalisation of both beta cell function and hepatic insulin sensitivity in type 2 diabetes was achieved by dietary energy restriction alone. This was associated with decreased pancreatic and liver triacylglycerol stores. The abnormalities underlying type 2 diabetes are reversible by reducing dietary energy intake.
Collapse
Affiliation(s)
- E. L. Lim
- Magnetic Resonance Centre, Institute of Cellular Medicine, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, NE4 5PL UK
| | - K. G. Hollingsworth
- Magnetic Resonance Centre, Institute of Cellular Medicine, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, NE4 5PL UK
| | - B. S. Aribisala
- Magnetic Resonance Centre, Institute of Cellular Medicine, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, NE4 5PL UK
| | - M. J. Chen
- Magnetic Resonance Centre, Institute of Cellular Medicine, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, NE4 5PL UK
| | - J. C. Mathers
- Human Nutrition Research Centre, Institute for Ageing and Health, Newcastle University, Newcastle upon Tyne, UK
| | - R. Taylor
- Magnetic Resonance Centre, Institute of Cellular Medicine, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, NE4 5PL UK
| |
Collapse
|
50
|
Abdul-Ghani MA, Norton L, Defronzo RA. Role of sodium-glucose cotransporter 2 (SGLT 2) inhibitors in the treatment of type 2 diabetes. Endocr Rev 2011; 32:515-31. [PMID: 21606218 DOI: 10.1210/er.2010-0029] [Citation(s) in RCA: 317] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Hyperglycemia plays an important role in the pathogenesis of type 2 diabetes mellitus, i.e., glucotoxicity, and it also is the major risk factor for microvascular complications. Thus, effective glycemic control will not only reduce the incidence of microvascular complications but also correct some of the metabolic abnormalities that contribute to the progression of the disease. Achieving durable tight glycemic control is challenging because of progressive β-cell failure and is hampered by increased frequency of side effects, e.g., hypoglycemia and weight gain. Most recently, inhibitors of the renal sodium-glucose cotransporter have been developed to produce glucosuria and reduce the plasma glucose concentration. These oral antidiabetic agents have the potential to improve glycemic control while avoiding hypoglycemia, to correct the glucotoxicity, and to promote weight loss. In this review, we will summarize the available data concerning the mechanism of action, efficacy, and safety of this novel antidiabetic therapeutic approach.
Collapse
|