1
|
Umano GR, Aiello F, Rondinelli G, Salvati A, Petrone D, Sibillo MV, D'Ausilio R, Marzuillo P, Grandone A, Cirillo G, Tricò D, Del Giudice EM. Obstructive sleep apnea is associated with impaired insulin clearance and hepatic insulin sensitivity in paediatric obesity. Diabetes Obes Metab 2025. [PMID: 40365648 DOI: 10.1111/dom.16464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/24/2025] [Accepted: 05/02/2025] [Indexed: 05/15/2025]
Abstract
AIMS Obstructive sleep apnea (OSA) affects up to 40% of children and adolescents with obesity and is linked to hyperinsulinism. However, the mechanisms underpinning this association remain unclear. The study aims to assess the three key determinants of hyperinsulinemia: insulin clearance, secretion, and sensitivity in paediatric patients with OSA. METHODS We enrolled 70 Children with obesity and suspected OSA who performed a nocturnal polygraphy to confirm OSA diagnosis and a 3-hour OGTT to assess glucose homeostasis. Mild OSA was defined for 1 < AHI ≤ 5, moderate-severe OSA as AHI >5. Insulin secretion rate (ISR) was estimated using C-peptide deconvolution. Basal and total insulin clearance during OGTT were calculated. Whole-body insulin sensitivity was evaluated with the Matsuda Index (WBISI), while hepatic insulin resistance index (HIRI) was calculated based on the AUCs of plasma glucose and insulin during the initial 30 minutes of OGTT. RESULTS A total of 36 children had mild OSA and 34 had moderate-severe OSA. The latter group showed reduced insulin clearance during OGTT (p = 0.008) and higher HIRI (p = 0.03). Basal insulin clearance (p = 0.07), ISR (p = 0.34), beta-cell glucose sensitivity (p = 0.53), and WBISI (p = 0.56) were similar between the two groups. OSA severity negatively correlated with fasting insulin clearance (r = -0.29, p = 0.01) and OGTT insulin clearance (r = -0.32, p = 0.007) and positively correlated with HIRI (r = 0.29, p = 0.02). CONCLUSION Moderate-severe OSA in children with obesity is associated with impaired insulin clearance and hepatic insulin sensitivity. These factors may contribute to hyperinsulinism in paediatric OSA.
Collapse
Affiliation(s)
- Giuseppina Rosaria Umano
- Department of the Woman, the Child, and General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Francesca Aiello
- Department of the Woman, the Child, and General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Giulia Rondinelli
- Department of the Woman, the Child, and General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Alessandra Salvati
- Department of the Woman, the Child, and General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Delfina Petrone
- Department of the Woman, the Child, and General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Maria Vittoria Sibillo
- Department of the Woman, the Child, and General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Raffaella D'Ausilio
- Department of the Woman, the Child, and General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Pierluigi Marzuillo
- Department of the Woman, the Child, and General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Anna Grandone
- Department of the Woman, the Child, and General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Grazia Cirillo
- Department of the Woman, the Child, and General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Domenico Tricò
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Emanuele Miraglia Del Giudice
- Department of the Woman, the Child, and General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
2
|
Eckstein Y, Kessler B, Hinrichs A, Novak I, von Thaden A, Lorenzen T, Rathkolb B, Scholz A, Blutke A, Koopmans SJ, de Angelis MH, Christoffersen BØ, Wolf E, Renner S. Protocol for in vivo assessment of glucose control and insulin secretion and sensitivity in the pig. STAR Protoc 2025; 6:103774. [PMID: 40249709 DOI: 10.1016/j.xpro.2025.103774] [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: 11/26/2024] [Revised: 02/03/2025] [Accepted: 03/26/2025] [Indexed: 04/20/2025] Open
Abstract
The pig is a valuable animal model in diabetes research; however, standardized protocols are essential for evaluating in vivo metabolism. Here, we present a protocol for in vivo assessment of glucose control and insulin secretion and sensitivity in the pig. We describe steps for catheter implantation, testing of intravenous glucose tolerance, performance of hyperinsulinemic-euglycemic clamps (HECs) and hyperglycemic clamps (HGCs), and blood processing. We then detail procedures for analysis of plasma glucose, insulin, glucagon, and C-peptide concentrations as well as data analysis. For complete details on the use and execution of this protocol, please refer to Renner et al.1 and Renner et al.2.
Collapse
Affiliation(s)
- Yasmin Eckstein
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, 81377 Munich, Germany; Center for Innovative Medical Models (CiMM), LMU Munich, 85764 Oberschleißheim, Germany
| | - Barbara Kessler
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, 81377 Munich, Germany; Center for Innovative Medical Models (CiMM), LMU Munich, 85764 Oberschleißheim, Germany
| | - Arne Hinrichs
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, 81377 Munich, Germany; Center for Innovative Medical Models (CiMM), LMU Munich, 85764 Oberschleißheim, Germany
| | - Istvan Novak
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, 81377 Munich, Germany; Center for Innovative Medical Models (CiMM), LMU Munich, 85764 Oberschleißheim, Germany
| | - Anne von Thaden
- German Center for Neurodegenerative Diseases (DZNE), LMU Munich, 81377 Munich, Germany
| | - Timo Lorenzen
- Institute of Veterinary Pathology at the Center for Clinical Veterinary Medicine, LMU Munich, 80539 Munich, Germany
| | - Birgit Rathkolb
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, 81377 Munich, Germany; German Mouse Clinic (GMC) and Genome Analysis Center (GAC), Institute of Experimental Genetics, Helmholtz Munich, 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Armin Scholz
- Livestock Center Oberschleissheim, LMU Munich, 85764 Oberschleißheim, Germany
| | - Andreas Blutke
- Institute of Veterinary Pathology at the Center for Clinical Veterinary Medicine, LMU Munich, 80539 Munich, Germany
| | - Sietse-Jan Koopmans
- Wageningen Livestock Research, Wageningen University & Research, Wageningen, the Netherlands
| | - Martin Hrabĕ de Angelis
- German Mouse Clinic (GMC) and Genome Analysis Center (GAC), Institute of Experimental Genetics, Helmholtz Munich, 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Chair of Experimental Genetics, TUM School of Life Sciences (SoLS), Technische Universität München, 85354 Freising, Germany
| | | | - Eckhard Wolf
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, 81377 Munich, Germany; Center for Innovative Medical Models (CiMM), LMU Munich, 85764 Oberschleißheim, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, 81377 Munich, Germany
| | - Simone Renner
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, 81377 Munich, Germany; Center for Innovative Medical Models (CiMM), LMU Munich, 85764 Oberschleißheim, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, 81377 Munich, Germany.
| |
Collapse
|
3
|
Rahman SMN, Yung JHM, Volchuk A, Goldenberg NM, Giacca A. Metabolic phenotypes in a Lyz2Cre recombinase mouse model. Front Immunol 2025; 16:1499858. [PMID: 40170862 PMCID: PMC11958952 DOI: 10.3389/fimmu.2025.1499858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 02/28/2025] [Indexed: 04/03/2025] Open
Abstract
The Cre-Lox system is essential in biomedical research for precise gene deletion in specific cell types, crucial for understanding genetic roles in disease. Although generally considered non-detrimental, Cre recombinase expression has been associated with potential adverse effects, including Cre toxicity, ectopic expression, and disruption of endogenous genes. We investigated the role of macrophage nucleotide-binding oligomerization domain (Nod1) in obesity-associated diabetes using myeloid-specific Nod1-knockout mice (Nod1 floxed crossed with Lyz2Cre). Our study examined Lyz2Cre as well as floxed control mice separately, unlike most research. Results indicated that Lyz2Cre expression alone impacts glucose metabolism, challenging the notion that Cre expression is harmless. This finding highlights the critical importance of including Cre-only controls in studies using floxed alleles to generate conditional knockout mouse models in order to ensure robust and accurate conclusions in molecular research.
Collapse
Affiliation(s)
- S. M. Niazur Rahman
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Justin Hou Ming Yung
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Allen Volchuk
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Neil M. Goldenberg
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Anesthesia and Pain Medicine, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Anesthesiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Adria Giacca
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Banting and Best Diabetes Centre, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
4
|
Galderisi A, Sims EK, Evans-Molina C, Petrelli A, Cuthbertson D, Nathan BM, Ismail HM, Herold KC, Moran A. Trajectory of beta cell function and insulin clearance in stage 2 type 1 diabetes: natural history and response to teplizumab. Diabetologia 2025; 68:646-661. [PMID: 39560746 PMCID: PMC11832608 DOI: 10.1007/s00125-024-06323-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 10/02/2024] [Indexed: 11/20/2024]
Abstract
AIMS/HYPOTHESIS We aimed to analyse TrialNet Anti-CD3 Prevention (TN10) data using oral minimal model (OMM)-derived indices to characterise the natural history of stage 2 type 1 diabetes in placebo-treated individuals, to describe early metabolic responses to teplizumab and to explore the predictive capacity of OMM measures for disease-free survival rate. METHODS OMM-estimated insulin secretion, sensitivity and clearance and the disposition index were evaluated at baseline and at 3, 6 and 12 months post randomisation in placebo- and teplizumab-treated groups, and, within each group, in slow- and rapid-progressors (time to stage 3 disease >2 or ≤ 2 years). OMM metrics were also compared with the standard AUC C-peptide. Percentage changes in CD8+ T memory cell and programmed death-1 (PD-1) expression were evaluated in each group. RESULTS Baseline metabolic characteristics were similar between 28 placebo- and 39 teplizumab-treated participants. Over 12 months, insulin secretion declined in placebo-treated and rose in teplizumab-treated participants. Within groups, placebo slow-progressors (n=14) maintained insulin secretion and sensitivity, while both declined in placebo rapid-progressors (n=14). Teplizumab slow-progressors (n=28) maintained elevated insulin secretion, while teplizumab rapid-progressors (n=11) experienced mild metabolic decline. Compared with rapid-progressor groups, insulin clearance significantly decreased between baseline and 3, 6 and 12 months in the slow-progressor groups in both treatment arms. In aggregate, both higher baseline insulin secretion (p=0.027) and reduced 12 month insulin clearance (p=0.045) predicted slower progression. A >25% loss of insulin secretion at 3 months had specificity of 0.95 (95% CI 0.86, 1.00) to identify rapid-progressors and correctly classified the 2 year risk for progression in 92% of participants, with a sensitivity of 0.19 (95% CI 0.08, 0.30). OMM-estimated insulin secretion outperformed AUC C-peptide to differentiate groups by treatment or to predict progression. Metabolic changes were paralleled by relative frequency of change in PD-1+ CD8+ T effector memory cells. CONCLUSIONS/INTERPRETATION OMM measures characterise the metabolic heterogeneity in stage 2 diabetes, identifying differences between rapid- and slow-progressors, and heterogeneous impacts of immunotherapy, suggesting the need to account for these differences when designing and interpreting clinical trials.
Collapse
Grants
- U01 DK061010 NIDDK NIH HHS
- UL1TR000142 Division of Diabetes, Endocrinology, and Metabolic Diseases
- UL1TR000445 Division of Diabetes, Endocrinology, and Metabolic Diseases
- R01 DK057846 NIDDK NIH HHS
- 3-SRA-2023-1422-S-B JDRF
- UC4 DK106993 NIDDK NIH HHS
- R01DK121929 Division of Diabetes, Endocrinology, and Metabolic Diseases
- U01 DK061042 NIDDK NIH HHS
- U01 DK085509 NIDDK NIH HHS
- DK106993 Division of Diabetes, Endocrinology, and Metabolic Diseases
- UL1 TR002366 NCATS NIH HHS
- UL1TR000064 Division of Diabetes, Endocrinology, and Metabolic Diseases
- U01 DK085476 NIDDK NIH HHS
- R01 DK133881 NIDDK NIH HHS
- R01 DK121929 NIDDK NIH HHS
- AI66387 Division of Diabetes, Endocrinology, and Metabolic Diseases
- DK057846 Division of Diabetes, Endocrinology, and Metabolic Diseases
- UL1 TR001857 NCATS NIH HHS
- UL1 TR000064 NCATS NIH HHS
- UL1 TR002537 NCATS NIH HHS
- U01 DK085466 NIDDK NIH HHS
- UL1 TR001872 NCATS NIH HHS
- U01 DK061058 NIDDK NIH HHS
- UL1 TR002529 NCATS NIH HHS
- UL1 TR001863 NCATS NIH HHS
- R01DK133881 Division of Diabetes, Endocrinology, and Metabolic Diseases
- U01 DK085453 NIDDK NIH HHS
- U01 DK106984 NIDDK NIH HHS
- UL1 TR000114 NCATS NIH HHS
- U01 DK085499 NIDDK NIH HHS
- U01 DK107013 NIDDK NIH HHS
- U01 DK103266 NIDDK NIH HHS
- F31 AI009565 NIAID NIH HHS
- K23 DK129799 NIDDK NIH HHS
- UM1 AI09565 Division of Diabetes, Endocrinology, and Metabolic Diseases
- K23DK129799 Division of Diabetes, Endocrinology, and Metabolic Diseases
- U01 DK103282 NIDDK NIH HHS
- 3-SRA-2022-1186-S-B JDRF
- 62288 John Templeton Foundation
- U01 DK107014 NIDDK NIH HHS
- U01 DK106994 NIDDK NIH HHS
- UL1 TR000142 NCATS NIH HHS
- U01 DK06 Division of Diabetes, Endocrinology, and Metabolic Diseases
- UL1TR002366 Division of Diabetes, Endocrinology, and Metabolic Diseases
- U01 DK061034 NIDDK NIH HHS
- UL1 TR001082 NCATS NIH HHS
- U01 DK085461 NIDDK NIH HHS
- UC4 DK097835 NIDDK NIH HHS
- U01 DK103180 NIDDK NIH HHS
- U01 DK085465 NIDDK NIH HHS
- U01 DK085504 NIDDK NIH HHS
Collapse
Affiliation(s)
| | - Emily K Sims
- Department of Pediatrics, Center for Diabetes and Metabolic Diseases, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Carmella Evans-Molina
- Department of Pediatrics, Center for Diabetes and Metabolic Diseases, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Alessandra Petrelli
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Pio Albergo Trivulzio, Milan, Italy
| | - David Cuthbertson
- Health Informatics Institute, University of South Florida, Tampa, FL, USA
| | - Brandon M Nathan
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Heba M Ismail
- Department of Pediatrics, Center for Diabetes and Metabolic Diseases, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kevan C Herold
- Department of Immunobiology, Yale University, New Haven, CT, USA
- Department of Internal Medicine, Yale University, New Haven, CT, USA
| | - Antoinette Moran
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
5
|
Nadeau KJ, Arslanian SA, Bacha F, Caprio S, Chao LC, Farrell R, Hughan KS, Rayas M, Tung M, Cross K, El Ghormli L. Insulin clearance at randomisation and in response to treatment in youth with type 2 diabetes: a secondary analysis of the TODAY randomised clinical trial. Diabetologia 2025; 68:676-687. [PMID: 39706874 DOI: 10.1007/s00125-024-06327-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 09/26/2024] [Indexed: 12/23/2024]
Abstract
AIMS/HYPOTHESIS Insulin resistance and compensatory hyperinsulinaemia are core features leading to beta cell failure in youth-onset type 2 diabetes. Insulin clearance (IC) is also a key regulator of insulin concentrations, but few data exist on IC in youth-onset type 2 diabetes. In a secondary analysis of our Treatment Options for Type 2 Diabetes in Adolescents and Youth (TODAY) randomised clinical trial, we investigated potential sex-, race-, ethnicity- and treatment-related differences in IC in youth-onset type 2 diabetes and aimed to identify metabolic phenotypes associated with IC at baseline and in response to metformin, metformin plus a lifestyle intervention, and metformin plus rosiglitazone. METHODS A total of 640 youth aged 10-18 years with type 2 diabetes underwent fasting blood tests, anthropometric measurements, dual-energy x-ray absorptiometry to estimate subcutaneous adipose tissue (SAT) and visceral adipose tissue (VAT) mass, and OGTTs longitudinally over 5 years. IC was calculated from the fasting C-peptide:insulin ratio (fasting IC) and 2 h OGTT C-peptide incremental AUC (iAUC):insulin iAUC ratio (2 h IC). Linear mixed models were used to assess covariate effects on the mean of IC over repeated time points. RESULTS Baseline fasting IC (×10-2 nmol/pmol) was significantly lower in female participants than male participants (median [IQR] 0.72 [0.57-0.93] vs 0.79 [0.63-1.00], respectively; p=0.04) and in non-Hispanic Black participants than Hispanic and non-Hispanic White participants (median [IQR] 0.64 [0.51-0.81] vs 0.78 [0.64-1.00] vs 0.84 [0.68-1.01], respectively; p<0.0001). Similar results were observed for 2 h IC. Lower IC most strongly correlated with higher weight over time (% change [95% CI] in IC per 5 kg increase: fasting IC -1.52 [-2.05, -0.99]; 2 h IC -3.46 [-4.05, -2.86]). Lower IC also correlated with other markers of adiposity (higher BMI and SAT mass), and markers of insulin sensitivity (higher waist:height ratio, VAT mass, VAT:SAT mass ratio, triacylglycerol concentrations, triacylglycerol:HDL-cholesterol ratio, aspartate aminotransferase [AST] and alanine aminotransferase [ALT] concentrations, and systolic and diastolic BP, and lower HDL-cholesterol and total and high molecular weight adiponectin concentrations) over time. Beta cell function as determined from OGTTs, not insulin sensitivity or IC, was predictive of persistently elevated blood glucose levels. IC was higher with metformin+rosiglitazone than metformin alone (p=0.03 for fasting IC; p=0.02 for 2 h IC) and metformin+lifestyle (2 h IC, p=0.005), but not after adjusting for adiponectin (p value not significant for all). CONCLUSIONS/INTERPRETATION In youth with type 2 diabetes, low IC is correlated with female sex, non-Hispanic Black race and ethnicity, and markers of adiposity and insulin resistance, but not with beta cell function. Along with insulin sensitivity and adiponectin, IC increased in response to rosiglitazone treatment. These findings suggest that, in youth-onset type 2 diabetes, low IC is a compensatory response to changes in insulin sensitivity and/or adiponectin concentrations and is not a mediator of beta cell function. TRIAL REGISTRATION ClinicalTrials.gov NCT00081328 DATA AVAILABILITY: Data from the TODAY study (V4; https://doi.org/10.58020/2w6w-pv88 ) reported here are available on request from the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) Central Repository (NIDDK-CR) Resources for Research ( https://repository.niddk.nih.gov/ ).
Collapse
Affiliation(s)
| | - Silva A Arslanian
- UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Fida Bacha
- Children's Nutrition Research Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Sonia Caprio
- Pediatric Endocrinology, Yale University School of Medicine, New Haven, CT, USA
| | - Lily C Chao
- Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Ryan Farrell
- University Hospitals Cleveland Medical Center, Rainbow Babies and Children's Hospital, Cleveland, OH, USA
| | - Kara S Hughan
- UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Maria Rayas
- University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Melinda Tung
- The Biostatistics Center, George Washington University, Rockville, MD, USA
| | - Kaitlyn Cross
- The Biostatistics Center, George Washington University, Rockville, MD, USA
| | - Laure El Ghormli
- The Biostatistics Center, George Washington University, Rockville, MD, USA.
| |
Collapse
|
6
|
Jiang L, Lai J, Xu X, Lu Y, Gu K, Chen S, Xu L, Liu K. Reduced insulin clearance in paediatric metabolic (dysfunction)-associated fatty liver disease and its dual role in beta-cell offload and diabetes risk. Diabetes Obes Metab 2024; 26:5390-5398. [PMID: 39192529 DOI: 10.1111/dom.15902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/31/2024] [Accepted: 08/10/2024] [Indexed: 08/29/2024]
Abstract
AIM Diminished hepatic insulin clearance (HIC) is observed in obese adults and is presumed to be mediated by fatty liver. However, few reports have examined HIC in Chinese children with metabolic (dysfunction)-associated fatty liver disease (MAFLD). This study aimed to investigate the correlation between HIC, insulin sensitivity and β-cell function in obese Chinese children with MAFLD. METHODS In total, 204 obese children (74 MAFLD) aged 4-17 years were enrolled into this study. HIC, insulin sensitivity and β-cell function were calculated using the oral glucose tolerance test (1.75 g/kg body weight). Correlation analyses between the HIC and clinical variables were performed using Pearson's product-moment correlation coefficients. HIC and glucose homeostasis were assessed in a high-fat diet mouse model, and liver samples were collected for molecular analysis. RESULTS Obese children with MAFLD exhibited significantly lower HIC (AUCC-peptide/insulin ratio, p = 0.0019), higher insulin resistance (homeostatic model assessment of insulin resistance, p = 0.002), and increased compensatory β-cell function (homeostatic model assessment-β, p = 0.046) than obese children without liver involvement. Notably, HIC was negatively correlated with insulin sensitivity (r = -0.5035, p < 0.0001) and β-cell function (r = -0.4576, p < 0.0001). However, pancreatic β-cell dysfunction (p = 0.046) was accompanied by future reduced HIC (p = 0.034) in children with MAFLD in prediabetes. In a high-fat diet mouse model, MAFLD mice showed a 50% reduction in insulin-degrading enzyme expression, consistent with the observed decrease in HIC. CONCLUSIONS A lower HIC may offload pancreatic β-cells at an early stage. However, obese children with MAFLD are at risk of developing diabetes, and preventive efforts should be prioritized.
Collapse
Affiliation(s)
- Li Jiang
- Department of Pediatric Endocrinology, Affiliated Children's Hospital of Jiangnan University (Wuxi Children's Hospital), Wuxi, China
| | - Jinxin Lai
- Department of Medical Laboratory, Wuxi Eighth People's Hospital, Wuxi, China
| | - Xu Xu
- Department of Pediatric Endocrinology, Affiliated Children's Hospital of Jiangnan University (Wuxi Children's Hospital), Wuxi, China
| | - Yang Lu
- Department of Pediatric Endocrinology, Affiliated Children's Hospital of Jiangnan University (Wuxi Children's Hospital), Wuxi, China
| | - Kefeng Gu
- Department of Pediatric Endocrinology, Affiliated Children's Hospital of Jiangnan University (Wuxi Children's Hospital), Wuxi, China
| | - Sha Chen
- Department of Obstetrics and Gynecology, Affiliated Women's Hospital of Jiangnan University (Wuxi Maternity and Child Health Care Hospital), Wuxi, China
| | - Lulian Xu
- Department of Pediatric Endocrinology, Affiliated Children's Hospital of Jiangnan University (Wuxi Children's Hospital), Wuxi, China
| | - Kerong Liu
- Department of Pediatric Endocrinology, Affiliated Children's Hospital of Jiangnan University (Wuxi Children's Hospital), Wuxi, China
| |
Collapse
|
7
|
Smith K, Taylor GS, Peeters W, Walker M, Perazzolo S, Atabaki-Pasdar N, Bowden Davies KA, Karpe F, Hodson L, Stevenson EJ, West DJ. Elevations in plasma glucagon are associated with reduced insulin clearance after ingestion of a mixed-macronutrient meal in people with and without type 2 diabetes. Diabetologia 2024; 67:2555-2567. [PMID: 39138690 PMCID: PMC11519192 DOI: 10.1007/s00125-024-06249-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 07/01/2024] [Indexed: 08/15/2024]
Abstract
AIMS/HYPOTHESIS The temporal suppression of insulin clearance after glucose ingestion is a key determinant of glucose tolerance for people without type 2 diabetes. Whether similar adaptations are observed after the ingestion of a mixed-macronutrient meal is unclear. METHODS In a secondary analysis of data derived from two randomised, controlled trials, we studied the temporal responses of insulin clearance after the ingestion of a standardised breakfast meal consisting of cereal and milk in lean normoglycaemic individuals (n=12; Lean-NGT), normoglycaemic individuals with central obesity (n=11; Obese-NGT) and in people with type 2 diabetes (n=19). Pre-hepatic insulin secretion rates were determined by the deconvolution of C-peptide, and insulin clearance was calculated using a single-pool model. Insulin sensitivity was measured by an oral minimal model. RESULTS There were divergent time course changes in insulin clearance between groups. In the Lean-NGT group, there was an immediate post-meal increase in insulin clearance compared with pre-meal values (p<0.05), whereas insulin clearance remained stable at baseline values in Obese-NGT or declined slightly in the type 2 diabetes group (p<0.05). The mean AUC for insulin clearance during the test was ~40% lower in the Obese-NGT (1.3 ± 0.4 l min-1 m-2) and type 2 diabetes (1.4 ± 0.7 l min-1 m-2) groups compared with Lean-NGT (1.9 ± 0.5 l min-1 m-2; p<0.01), with no difference between the Obese-NGT and type 2 diabetes groups. HOMA-IR and glucagon AUC emerged as predictors of insulin clearance AUC, independent of BMI, age or insulin sensitivity (adjusted R2=0.670). Individuals with increased glucagon AUC had a 40% reduction in insulin clearance AUC (~ -0.75 l min-1 m-2; p<0.001). CONCLUSIONS/INTERPRETATION The ingestion of a mixed-macronutrient meal augments differing temporal profiles in insulin clearance among individuals without type 2 diabetes, which is associated with HOMA-IR and the secretion of glucagon. Further research investigating the role of hepatic glucagon signalling in postprandial insulin kinetics is warranted. TRIAL REGISTRATION ISRCTN17563146 and ISRCTN95281775.
Collapse
Affiliation(s)
- Kieran Smith
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, University of Oxford, Oxford, UK.
- Human Nutrition and Exercise Research Centre, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK.
- School of Biomedical, Nutritional, and Sport Sciences, Newcastle University, Newcastle upon Tyne, UK.
| | - Guy S Taylor
- Human Nutrition and Exercise Research Centre, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
- School of Biomedical, Nutritional, and Sport Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Wouter Peeters
- Human Nutrition and Exercise Research Centre, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
- School of Biomedical, Nutritional, and Sport Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Mark Walker
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Simone Perazzolo
- Nanomath LLC, Spokane, WA, USA
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| | - Naeimeh Atabaki-Pasdar
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospital Trust, Oxford, UK
- Genetic and Molecular Epidemiology Unit, Lund University Diabetes Centre, Department of Clinical Science, Lund University, Malmö, Sweden
| | - Kelly A Bowden Davies
- Human Nutrition and Exercise Research Centre, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
- Sport and Exercise Sciences, Manchester Metropolitan University, Manchester, UK
| | - Fredrik Karpe
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospital Trust, Oxford, UK
| | - Leanne Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospital Trust, Oxford, UK
| | - Emma J Stevenson
- Human Nutrition and Exercise Research Centre, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
- School of Biomedical, Nutritional, and Sport Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Daniel J West
- Human Nutrition and Exercise Research Centre, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK.
- School of Biomedical, Nutritional, and Sport Sciences, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
8
|
Kjeldsen SAS, Richter MM, Jensen NJ, Nilsson MSD, Heinz N, Nybing JD, Linden FH, Høgh-Schmidt E, Boesen MP, Andersen TL, Johannesen HH, Trammell SAJ, Grevengoed TJ, Madsbad S, Vilstrup H, Schiødt FV, Møller A, Rashu EB, Nørgaard K, Schmidt S, Gluud LL, Haugaard SB, Holst JJ, Rungby J, Wewer Albrechtsen NJ. Glucagon Resistance in Individuals With Obesity and Hepatic Steatosis Can Be Measured Using the GLUSENTIC Test and Index. Diabetes 2024; 73:1716-1727. [PMID: 38976454 DOI: 10.2337/db23-0858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 06/21/2024] [Indexed: 07/10/2024]
Abstract
Increased plasma levels of glucagon (hyperglucagonemia) promote diabetes development but are also observed in patients with metabolic dysfunction-associated steatotic liver disease (MASLD). This may reflect hepatic glucagon resistance toward amino acid catabolism. A clinical test for measuring glucagon resistance has not been validated. We evaluated our glucagon sensitivity (GLUSENTIC) test, which consists of 2 study days: a glucagon injection and measurements of plasma amino acids and an infusion of mixed amino acids and subsequent calculation of the GLUSENTIC index (primary outcome measure) from measurements of glucagon and amino acids. To distinguish glucagon-dependent from insulin-dependent actions on amino acid metabolism, we also studied patients with type 1 diabetes (T1D). The δ-decline in total amino acids was 49% lower in MASLD following exogenous glucagon (P = 0.01), and the calculated GLUSENTIC index was 34% lower in MASLD (P < 0.0001) but not T1D (P > 0.99). In contrast, glucagon-induced glucose increments were similar in control participants and participants with MASLD (P = 0.41). The GLUSENTIC test and index may be used to measure glucagon resistance in individuals with obesity and MASLD. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Sasha A S Kjeldsen
- Department of Clinical Biochemistry, Copenhagen University Hospital Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael M Richter
- Department of Clinical Biochemistry, Copenhagen University Hospital Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nicole J Jensen
- Department of Endocrinology, Copenhagen University Hospital Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Malin S D Nilsson
- Department of Endocrinology, Copenhagen University Hospital Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Niklas Heinz
- Department of Clinical Biochemistry, Copenhagen University Hospital Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Janus D Nybing
- Department of Radiology, Copenhagen University Hospital Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Frederik H Linden
- Department of Radiology, Copenhagen University Hospital Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Erik Høgh-Schmidt
- Department of Radiology, Copenhagen University Hospital Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Mikael P Boesen
- Department of Radiology, Copenhagen University Hospital Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas L Andersen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Helle H Johannesen
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Samuel A J Trammell
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Trisha J Grevengoed
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sten Madsbad
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Endocrinology, Hvidovre University Hospital, Hvidovre, Denmark
| | - Hendrik Vilstrup
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
| | - Frank Vinholt Schiødt
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Digestive Disease Center K, Copenhagen University Hospital Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Andreas Møller
- Gastro Unit, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Elias B Rashu
- Gastro Unit, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Kirsten Nørgaard
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Steno Diabetes Center Copenhagen, Herlev, Denmark
| | | | - Lise L Gluud
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Gastro Unit, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Steen B Haugaard
- Department of Endocrinology, Copenhagen University Hospital Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jørgen Rungby
- Department of Endocrinology, Copenhagen University Hospital Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Nicolai J Wewer Albrechtsen
- Department of Clinical Biochemistry, Copenhagen University Hospital Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
9
|
Agrinier AL, Morissette A, Daoust L, Gignac T, Marois J, Varin TV, Pilon G, Larose É, Gagnon C, Desjardins Y, Anhê FF, Carreau AM, Vohl MC, Marette A. Camu-camu decreases hepatic steatosis and liver injury markers in overweight, hypertriglyceridemic individuals: A randomized crossover trial. Cell Rep Med 2024; 5:101682. [PMID: 39168095 PMCID: PMC11384942 DOI: 10.1016/j.xcrm.2024.101682] [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: 03/20/2023] [Revised: 04/16/2024] [Accepted: 07/18/2024] [Indexed: 08/23/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) affects 25% of the adult population with no effective drug treatments available. Previous animal studies reported that a polyphenol-rich extract from the Amazonian berry camu-camu (CC) prevented hepatic steatosis in a mouse model of diet-induced obesity. This study aims to determine the impact of CC on hepatic steatosis (primary outcome) and evaluate changes in metabolic and gut microbiota profiles (exploratory outcomes). A randomized, double-blind, placebo-controlled crossover trial is conducted on 30 adults with overweight and hypertriglyceridemia, who consume 1.5 g of CC capsules or placebo daily for 12 weeks. CC treatment decreases liver fat by 7.43%, while it increases by 8.42% during the placebo intervention, showing a significant difference of 15.85%. CC decreases plasma aspartate and alanine aminotransferases levels and promotes changes in gut microbiota composition. These findings support that polyphenol-rich prebiotic may reduce liver fat in adults with overweight, reducing the risk of developing NAFLD.
Collapse
Affiliation(s)
- Anne-Laure Agrinier
- Department of Medicine, Faculty of Medicine, Québec Heart and Lung Institute (IUCPQ), Université Laval, Quebec City, QC, Canada; Centre Nutrition, santé et société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, QC, Canada
| | - Arianne Morissette
- Department of Medicine, Faculty of Medicine, Québec Heart and Lung Institute (IUCPQ), Université Laval, Quebec City, QC, Canada; Centre Nutrition, santé et société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, QC, Canada
| | - Laurence Daoust
- Department of Medicine, Faculty of Medicine, Québec Heart and Lung Institute (IUCPQ), Université Laval, Quebec City, QC, Canada; Centre Nutrition, santé et société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, QC, Canada
| | - Théo Gignac
- Department of Medicine, Faculty of Medicine, Centre de Recherche CHU de Québec-Université Laval, Université Laval, Quebec City, QC, Canada
| | - Julie Marois
- Centre Nutrition, santé et société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, QC, Canada
| | - Thibault V Varin
- Centre Nutrition, santé et société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, QC, Canada
| | - Geneviève Pilon
- Department of Medicine, Faculty of Medicine, Québec Heart and Lung Institute (IUCPQ), Université Laval, Quebec City, QC, Canada; Centre Nutrition, santé et société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, QC, Canada
| | - Éric Larose
- Department of Medicine, Faculty of Medicine, Québec Heart and Lung Institute (IUCPQ), Université Laval, Quebec City, QC, Canada
| | - Claudia Gagnon
- Department of Medicine, Faculty of Medicine, Québec Heart and Lung Institute (IUCPQ), Université Laval, Quebec City, QC, Canada; Department of Medicine, Faculty of Medicine, Centre de Recherche CHU de Québec-Université Laval, Université Laval, Quebec City, QC, Canada
| | - Yves Desjardins
- Centre Nutrition, santé et société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, QC, Canada; Department of Plant Science, Faculty of Agriculture and Food sciences, Université Laval, Quebec City, QC, Canada
| | - Fernando F Anhê
- Department of Medicine, Faculty of Medicine, Québec Heart and Lung Institute (IUCPQ), Université Laval, Quebec City, QC, Canada; Centre Nutrition, santé et société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, QC, Canada
| | - Anne-Marie Carreau
- Department of Medicine, Faculty of Medicine, Centre de Recherche CHU de Québec-Université Laval, Université Laval, Quebec City, QC, Canada
| | - Marie-Claude Vohl
- Centre Nutrition, santé et société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, QC, Canada; School of Nutrition, Université Laval, Quebec City, QC, Canada
| | - André Marette
- Department of Medicine, Faculty of Medicine, Québec Heart and Lung Institute (IUCPQ), Université Laval, Quebec City, QC, Canada; Centre Nutrition, santé et société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, QC, Canada.
| |
Collapse
|
10
|
Fan Y, Chow E, Lim CKP, Hou Y, Tsoi STF, Fan B, Lau ESH, Kong APS, Ma RCW, Wu H, Chan JCN, Luk AOY. Comparison of β-Cell Function and Insulin Sensitivity Between Normal-Weight and Obese Chinese With Young-Onset Type 2 Diabetes. Diabetes 2024; 73:953-963. [PMID: 38506952 DOI: 10.2337/db23-0966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 03/17/2024] [Indexed: 03/22/2024]
Abstract
Normal-weight individuals with usual-onset type 2 diabetes have reduced β-cell function and greater insulin sensitivity compared with their obese counterparts. The relative contribution of β-cell dysfunction and insulin resistance to young-onset type 2 diabetes (YOD) among normal-weight individuals is not well established. In 44 individuals with YOD (24 with normal weight and 20 with obesity) and 24 healthy control individuals with normoglycemia (12 with normal weight and 12 with obesity), we conducted 2-h 12 mmol/L hyperglycemic clamps to measure acute (0-10 min) and steady-state (100-120 min) insulin and C-peptide responses, as well as insulin sensitivity index. Normal-weight individuals with YOD had lower acute insulin response, steady-state insulin and C-peptide responses, and a higher insulin sensitivity index compared with their obese counterparts with YOD. Compared with BMI-matched healthy control individuals, normal-weight individuals with YOD had lower acute and steady-state insulin and C-peptide responses but a similar insulin sensitivity index. The impairment of steady-state β-cell response relative to healthy control individuals was more pronounced in normal-weight versus obese individuals with YOD. In conclusion, normal-weight Chinese with YOD exhibited worse β-cell function but preserved insulin sensitivity relative to obese individuals with YOD and BMI-matched healthy individuals with normoglycemia. The selection of glucose-lowering therapy should account for pathophysiological differences underlying YOD between normal-weight and obese individuals. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Yingnan Fan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Elaine Chow
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Phase 1 Clinical Trial Centre, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Cadmon K P Lim
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Yong Hou
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Sandra T F Tsoi
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Baoqi Fan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Eric S H Lau
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Alice P S Kong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Ronald C W Ma
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Hongjiang Wu
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Juliana C N Chan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Andrea O Y Luk
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Phase 1 Clinical Trial Centre, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| |
Collapse
|
11
|
Jensen ASH, Ytting H, Werge MP, Rashu EB, Hetland LE, Thing M, Nabilou P, Burisch J, Bojsen-Møller KN, Junker AE, Hobolth L, Mortensen C, Tofteng F, Bendtsen F, Møller S, Vyberg M, Serizawa RR, Gluud LL, Wewer Albrechtsen NJ. Patients with autoimmune liver disease have glucose disturbances that mechanistically differ from steatotic liver disease. Am J Physiol Gastrointest Liver Physiol 2024; 326:G736-G746. [PMID: 38625142 DOI: 10.1152/ajpgi.00047.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 03/25/2024] [Accepted: 04/09/2024] [Indexed: 04/17/2024]
Abstract
Autoimmune liver diseases are associated with an increased risk of diabetes, yet the underlying mechanisms remain unknown. In this cross-sectional study, we investigated the glucose-regulatory disturbances in patients with autoimmune hepatitis (AIH, n = 19), primary biliary cholangitis (PBC, n = 15), and primary sclerosing cholangitis (PSC, n = 6). Healthy individuals (n = 24) and patients with metabolic dysfunction-associated steatotic liver disease (MASLD, n = 18) were included as controls. Blood samples were collected during a 120-min oral glucose tolerance test. We measured the concentrations of glucose, C-peptide, insulin, glucagon, and the two incretin hormones, glucose insulinotropic peptide (GIP) and glucagon-like peptide-1 (GLP-1). We calculated the homeostasis model assessment of insulin resistance (HOMA-IR), whole body insulin resistance (Matsuda index), insulin clearance, and insulinogenic index. All patient groups had increased fasting plasma glucose and impaired glucose responses compared with healthy controls. Beta-cell secretion was increased in AIH, PBC, and MASLD but not in PSC. Patients with AIH and MASLD had hyperglucagonemia and hepatic, as well as peripheral, insulin resistance and decreased insulin clearance, resulting in hyperinsulinemia. Patients with autoimmune liver disease had an increased GIP response, and those with AIH or PBC had an increased GLP-1 response. Our data demonstrate that the mechanism underlying glucose disturbances in patients with autoimmune liver disease differs from that underlying MASLD, including compensatory incretin responses in patients with autoimmune liver disease. Our results suggest that glucose disturbances are present at an early stage of the disease.NEW & NOTEWORTHY Patients with autoimmune liver disease but without overt diabetes display glucose disturbances early on in their disease course. We identified pathophysiological traits specific to these patients including altered incretin responses.
Collapse
Affiliation(s)
- Anne-Sofie H Jensen
- Gastro Unit, Copenhagen University Hospital-Amager and Hvidovre Hospital, Hvidovre, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital-Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Henriette Ytting
- Gastro Unit, Copenhagen University Hospital-Amager and Hvidovre Hospital, Hvidovre, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mikkel P Werge
- Gastro Unit, Copenhagen University Hospital-Amager and Hvidovre Hospital, Hvidovre, Denmark
| | - Elias B Rashu
- Gastro Unit, Copenhagen University Hospital-Amager and Hvidovre Hospital, Hvidovre, Denmark
| | - Liv E Hetland
- Gastro Unit, Copenhagen University Hospital-Amager and Hvidovre Hospital, Hvidovre, Denmark
| | - Mira Thing
- Gastro Unit, Copenhagen University Hospital-Amager and Hvidovre Hospital, Hvidovre, Denmark
| | - Puria Nabilou
- Gastro Unit, Copenhagen University Hospital-Amager and Hvidovre Hospital, Hvidovre, Denmark
| | - Johan Burisch
- Gastro Unit, Copenhagen University Hospital-Amager and Hvidovre Hospital, Hvidovre, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kirstine N Bojsen-Møller
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Endocrinology, Copenhagen University Hospital-Amager and Hvidovre Hospital, Hvidovre, Denmark
| | - Anders E Junker
- Gastro Unit, Copenhagen University Hospital-Amager and Hvidovre Hospital, Hvidovre, Denmark
| | - Lise Hobolth
- Gastro Unit, Copenhagen University Hospital-Amager and Hvidovre Hospital, Hvidovre, Denmark
| | - Christian Mortensen
- Gastro Unit, Copenhagen University Hospital-Amager and Hvidovre Hospital, Hvidovre, Denmark
| | - Flemming Tofteng
- Gastro Unit, Copenhagen University Hospital-Amager and Hvidovre Hospital, Hvidovre, Denmark
| | - Flemming Bendtsen
- Gastro Unit, Copenhagen University Hospital-Amager and Hvidovre Hospital, Hvidovre, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Søren Møller
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Physiology and Nuclear Medicine, Center for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital-Amager and Hvidovre Hospital, Hvidovre, Denmark
| | - Mogens Vyberg
- Department of Pathology, Copenhagen University Hospital-Amager and Hvidovre Hospital, Hvidovre, Denmark
- Department of Clinical Medicine, Center for RNA Medicine, Aalborg University, Copenhagen, Denmark
| | - Reza R Serizawa
- Department of Pathology, Copenhagen University Hospital-Amager and Hvidovre Hospital, Hvidovre, Denmark
| | - Lise L Gluud
- Gastro Unit, Copenhagen University Hospital-Amager and Hvidovre Hospital, Hvidovre, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nicolai J Wewer Albrechtsen
- Department of Clinical Biochemistry, Copenhagen University Hospital-Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
12
|
Yau B, Madsen S, Nelson ME, Cooke KC, Fritzen AM, Thorius IH, Stöckli J, James DE, Kebede MA. Genetics and diet shape the relationship between islet function and whole body metabolism. Am J Physiol Endocrinol Metab 2024; 326:E663-E672. [PMID: 38568150 PMCID: PMC11376487 DOI: 10.1152/ajpendo.00060.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/28/2024] [Accepted: 03/30/2024] [Indexed: 05/08/2024]
Abstract
Despite the fact that genes and the environment are known to play a central role in islet function, our knowledge of how these parameters interact to modulate insulin secretory function remains relatively poor. Presently, we performed ex vivo glucose-stimulated insulin secretion and insulin content assays in islets of 213 mice from 13 inbred mouse strains on chow, Western diet (WD), and a high-fat, carbohydrate-free (KETO) diet. Strikingly, among these 13 strains, islets from the commonly used C57BL/6J mouse strain were the least glucose responsive. Using matched metabolic phenotyping data, we performed correlation analyses of isolated islet parameters and found a positive correlation between basal and glucose-stimulated insulin secretion, but no relationship between insulin secretion and insulin content. Using in vivo metabolic measures, we found that glucose tolerance determines the relationship between ex vivo islet insulin secretion and plasma insulin levels. Finally, we showed that islet glucose-stimulated insulin secretion decreased with KETO in almost all strains, concomitant with broader phenotypic changes, such as increased adiposity and glucose intolerance. This is an important finding as it should caution against the application of KETO diet for beta-cell health. Together these data offer key insights into the intersection of diet and genetic background on islet function and whole body glucose metabolism.NEW & NOTEWORTHY Thirteen strains of mice on chow, Western diet, and high-fat, carbohydrate-free (KETO), correlating whole body phenotypes to ex vivo pancreatic islet functional measurements, were used. The study finds a huge spectrum of functional islet responses and insulin phenotypes across all strains and diets, with the ubiquitous C57Bl/6J mouse exhibiting the lowest secretory response of all strains, highlighting the overall importance of considering genetic background when investigating islet function. Ex vivo basal and stimulated insulin secretion are correlated in the islet, and KETO imparts widescale downregulation of islet insulin secretion.
Collapse
Affiliation(s)
- Belinda Yau
- School of Medical Science, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Søren Madsen
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia
| | - Marin E Nelson
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia
| | - Kristen C Cooke
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia
| | - Andreas M Fritzen
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia
| | - Ida H Thorius
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia
| | - Jacqueline Stöckli
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia
| | - David E James
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia
- Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia
| | - Melkam A Kebede
- School of Medical Science, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| |
Collapse
|
13
|
Godsland IF, Johnston DG, Alberti K, Oliver N. The importance of intravenous glucose tolerance test glucose stimulus for the evaluation of insulin secretion. Sci Rep 2024; 14:7451. [PMID: 38548796 PMCID: PMC10978989 DOI: 10.1038/s41598-024-54584-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 02/14/2024] [Indexed: 04/01/2024] Open
Abstract
For 100 years, the Intravenous glucose tolerance test (IVGTT) has been used extensively in researching the pathophysiology of diabetes mellitus and AIRg-the IVGTT-induced acute insulin response to the rapid rise in circulating glucose-is a key measure of insulin secretory capacity. For an effective evaluation of AIRg, IVGTT glucose loading should be adjusted for glucose distribution volume (gVOL) to provide an invariant, trend-free immediate rise in circulating glucose (ΔG0). Body weight-based glucose loads have been widely used but whether these achieve a trend-free ΔG0 does not appear to have been investigated. By analysing variation in AIRg, ΔG0 and gVOL with a range of IVGTT loads, both observed and simulated, we explored the hypothesis that there would be an optimum anthropometry-based IVGTT load calculation that, by achieving a trend-free ΔG0, would not compromise evaluation of AIRg as an index of beta cell function. Data derived from patient and research volunteer records for 3806 IVGTT glucose and insulin profiles. Among the non-obese, as gVOL rose, weight increased disproportionately rapidly. Consequently, the IVGTT glucose load needed for an invariant ΔG0 was progressively overestimated, accounting for 47% of variation in AIRg. Among the obese, ΔG0 was trend-free yet AIRg increased by 11.6% per unit body mass index, consistent with a more proportionate increase in weight with gVOL and a hyperinsulinaemic adaptation to adiposity-associated insulin resistance. Simulations further confirmed our hypothesis by demonstrating that a body surface area-based IVGTT load calculation could provide for a more generally invariant IVGTT ΔG0.
Collapse
Affiliation(s)
- Ian F Godsland
- Division of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK.
- Wynn Reader in Human Metabolism, Section of Metabolic Medicine, Faculty of Medicine, Imperial College London (St Mary's Campus), Room G1, Norfolk Place, London, W2 2NH, UK.
| | - Desmond G Johnston
- Division of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
- Department of Diabetes and Endocrinology, St Marys Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Kgmm Alberti
- Division of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | - Nick Oliver
- Division of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
- Department of Diabetes and Endocrinology, St Marys Hospital, Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
14
|
Suppli MP, Høgedal A, Bagger JI, Chabanova E, van Hall G, Forman JL, Christensen MB, Albrechtsen NJW, Holst JJ, Knop FK. Signs of Glucagon Resistance After a 2-Week Hypercaloric Diet Intervention. J Clin Endocrinol Metab 2024; 109:955-967. [PMID: 37967235 DOI: 10.1210/clinem/dgad666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/06/2023] [Accepted: 11/13/2023] [Indexed: 11/17/2023]
Abstract
CONTEXT Hyperglucagonemia is observed in individuals with obesity and contributes to the hyperglycemia of patients with type 2 diabetes. Hyperglucagonemia may develop due to steatosis-induced hepatic glucagon resistance resulting in impaired hepatic amino acid turnover and ensuing elevations of circulating glucagonotropic amino acids. OBJECTIVE We evaluated whether glucagon resistance could be induced in healthy individuals by a hypercaloric diet intervention designed to increase hepatic fat content. METHODS We recruited 20 healthy male individuals to follow a hypercaloric diet and a sedentary lifestyle for 2 weeks. Amino acid concentrations in response to infusion of glucagon were assessed during a pancreatic clamp with somatostatin and basal insulin. The reversibility of any metabolic changes was assessed 8 weeks after the intervention. Hepatic steatosis was assessed by magnetic resonance spectroscopy. RESULTS The intervention led to increased hepatic fat content (382% [206%; 705%], P < .01). Glucagon infusion led to a decrease in the concentration of total amino acids on all experimental days, but the percentage change in total amino acids was reduced (-2.5% ± 0.5% vs -0.2% ± 0.7%, P = .015) and the average slope of the decline in the total amino acid concentration was less steep (-2.0 ± 1.2 vs -1.2 ± 0.3 μM/min, P = .016) after the intervention compared to baseline. The changes were normalized at follow-up. CONCLUSION Our results indicate that short-term unhealthy behavior, which increases hepatic fat content, causes a reversible resistance to the effect of glucagon on amino acid concentrations in healthy individuals, which may explain the hyperglucagonemia associated with obesity and diabetes.
Collapse
Affiliation(s)
- Malte Palm Suppli
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
| | - Astrid Høgedal
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
| | - Jonatan Ising Bagger
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
- Steno Diabetes Center Copenhagen, DK-2730 Herlev, Denmark
| | - Elizaveta Chabanova
- Department of Radiology, Herlev Hospital, University of Copenhagen, DK-2730 Herlev, Denmark
| | - Gerrit van Hall
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
- Clinical Metabolomics Core Facility, Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Julie Lyng Forman
- Section of Biostatistics, Department of Public Health, University of Copenhagen, DK-1353 Copenhagen, Denmark
| | - Mikkel Bring Christensen
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
- Department of Clinical Pharmacology, Bispebjerg Hospital, University of Copenhagen, DK-2400 Copenhagen, Denmark
- Copenhagen Center for Translational Research, Bispebjerg Hospital, University of Copenhagen, DK-2400 Copenhagen, Denmark
| | - Nicolai Jacob Wewer Albrechtsen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
- Department of Clinical Biochemistry, Bispebjerg and Frederiksberg Hospital, University of Copenhagen, DK-2400 Copenhagen, Denmark
| | - Jens Juul Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Filip Krag Knop
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
- Steno Diabetes Center Copenhagen, DK-2730 Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| |
Collapse
|
15
|
Mak AL, Wassenaar N, van Dijk AM, Troelstra M, Houttu V, van Son K, Driessen S, Zwirs D, van den Berg-Faay S, Shumbayawonda E, Runge J, Doukas M, Verheij J, Beuers U, Nieuwdorp M, Cahen DL, Nederveen A, Gurney-Champion O, Holleboom A. Intrapancreatic fat deposition is unrelated to liver steatosis in metabolic dysfunction-associated steatotic liver disease. JHEP Rep 2024; 6:100998. [PMID: 38379586 PMCID: PMC10877191 DOI: 10.1016/j.jhepr.2023.100998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 11/21/2023] [Accepted: 12/21/2023] [Indexed: 02/22/2024] Open
Abstract
Background & Aims Individuals with obesity may develop intrapancreatic fat deposition (IPFD) and fatty pancreas disease (FPD). Whether this causes inflammation and fibrosis and leads to pancreatic dysfunction is less established than for liver damage in metabolic dysfunction-associated steatotic liver disease (MASLD). Moreover, the interrelations of FPD and MASLD are poorly understood. Therefore, we aimed to assess IPFD and fibro-inflammation in relation to pancreatic function and liver disease severity in individuals with MASLD. Methods Seventy-six participants from the Amsterdam MASLD-MASH cohort (ANCHOR) study underwent liver biopsy and multiparametric MRI of the liver and pancreas, consisting of proton-density fat fraction sequences, T1 mapping and intravoxel incoherent motion diffusion-weighted imaging (IVIM-DWI). Results The prevalence of FPD was 37.3%. There was a clear correlation between pancreatic T1 relaxation time, which indicates fibro-inflammation, and parameters of glycemic dysregulation, namely HbA1c (R = 0.59; p <0.001), fasting glucose (R = 0.51; p <0.001) and the presence of type 2 diabetes (mean 802.0 ms vs. 733.6 ms; p <0.05). In contrast, there was no relation between IPFD and hepatic fat content (R = 0.03; p = 0.80). Pancreatic IVIM diffusion (IVIM-D) was lower in advanced liver fibrosis (p <0.05) and pancreatic perfusion (IVIM-f), reflecting vessel density, inversely correlated to histological MASLD activity (p <0.05). Conclusions Consistent relations exist between pancreatic fibro-inflammation on MRI and endocrine function in individuals with MASLD. However, despite shared dysmetabolic drivers, our study suggests IPFD is a separate pathophysiological process from MASLD. Impact and implications Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common chronic liver disease worldwide and 68% of people with type 2 diabetes have MASLD. However, fat infiltration and inflammation in the pancreas are understudied in individuals with MASLD. In this cross-sectional MRI study, we found no relationship between fat accumulation in the pancreas and liver in a cohort of patients with MASLD. However, our results show that inflammatory and fibrotic processes in the pancreas may be interrelated to features of type 2 diabetes and to the severity of liver disease in patients with MASLD. Overall, the results suggest that pancreatic endocrine dysfunction in individuals with MASLD may be more related to glucotoxicity than to lipotoxicity. Clinical trial number NTR7191 (Dutch Trial Register).
Collapse
Affiliation(s)
- Anne Linde Mak
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Nienke Wassenaar
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Anne-Marieke van Dijk
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Marian Troelstra
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Veera Houttu
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Koen van Son
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Gastroenterology and Hepatology, Radboudumc, Nijmegen, The Netherlands
| | - Stan Driessen
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Diona Zwirs
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Sandra van den Berg-Faay
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | | | - Jurgen Runge
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Michail Doukas
- Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Joanne Verheij
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Pathology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Ulrich Beuers
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Max Nieuwdorp
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Djuna L. Cahen
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Aart Nederveen
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Oliver Gurney-Champion
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Adriaan Holleboom
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
16
|
Gray SM, Goonatilleke E, Emrick MA, Becker JO, Hoofnagle AN, Stefanovski D, He W, Zhang G, Tong J, Campbell J, D’Alessio DA. High Doses of Exogenous Glucagon Stimulate Insulin Secretion and Reduce Insulin Clearance in Healthy Humans. Diabetes 2024; 73:412-425. [PMID: 38015721 PMCID: PMC10882148 DOI: 10.2337/db23-0201] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 11/20/2023] [Indexed: 11/30/2023]
Abstract
Glucagon is generally defined as a counterregulatory hormone with a primary role to raise blood glucose concentrations by increasing endogenous glucose production (EGP) in response to hypoglycemia. However, glucagon has long been known to stimulate insulin release, and recent preclinical findings have supported a paracrine action of glucagon directly on islet β-cells that augments their secretion. In mice, the insulinotropic effect of glucagon is glucose dependent and not present during basal euglycemia. To test the hypothesis that the relative effects of glucagon on hepatic and islet function also vary with blood glucose, a group of healthy subjects received glucagon (100 ng/kg) during fasting glycemia or experimental hyperglycemia (∼150 mg/dL) on 2 separate days. During fasting euglycemia, administration of glucagon caused blood glucose to rise due to increased EGP, with a delayed increase of insulin secretion. When given during experimental hyperglycemia, glucagon caused a rapid, threefold increase in insulin secretion, as well as a more gradual increase in EGP. Under both conditions, insulin clearance was decreased in response to glucagon infusion. The insulinotropic action of glucagon, which is proportional to the degree of blood glucose elevation, suggests distinct physiologic roles in the fasting and prandial states. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Sarah M. Gray
- Duke Molecular Physiology Institute, Duke University, Durham, NC
| | - Elisha Goonatilleke
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA
| | - Michelle A. Emrick
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA
| | - Jessica O. Becker
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA
| | - Andrew N. Hoofnagle
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA
| | - Darko Stefanovski
- Department of Clinical Studies–New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Kennett Square
| | - Wentao He
- Duke Molecular Physiology Institute, Duke University, Durham, NC
| | - Guofang Zhang
- Duke Molecular Physiology Institute, Duke University, Durham, NC
| | - Jenny Tong
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA
- Endocrine Section, VA Puget Sound Health Care System, Seattle
| | - Jonathan Campbell
- Duke Molecular Physiology Institute, Duke University, Durham, NC
- Division of Endocrinology, Department of Medicine, Duke University, Durham, NC
| | - David A. D’Alessio
- Duke Molecular Physiology Institute, Duke University, Durham, NC
- Division of Endocrinology, Department of Medicine, Duke University, Durham, NC
| |
Collapse
|
17
|
Ang T, Mason SA, Dao GM, Bruce CR, Kowalski GM. The impact of a single dose of whey protein on glucose flux and metabolite profiles in normoglycemic males: insights into glucagon and insulin biology. Am J Physiol Endocrinol Metab 2023; 325:E688-E699. [PMID: 37877796 DOI: 10.1152/ajpendo.00182.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/26/2023]
Abstract
Protein ingestion concurrently stimulates euglycemic glucagon and insulin secretion, a response that is particularly robust with rapidly absorbing proteins. Previously, we have shown that ingestion of repeated doses of rapidly absorbing whey protein equally stimulated endogenous glucose production (EGP) and glucose disposal (Rd), thus explaining the preservation of euglycemia. Here, we aimed to determine if a smaller single dose of whey could elicit a large enough glucagon and insulin response to stimulate glucose flux. Therefore, in normoglycemic young adult males (n = 10; age ∼26; BMI ∼25), using [6,6-2H2] glucose tracing and quantitative targeted metabolite profiling, we determined the metabolic response to a single 25 g "standard" dose of whey protein. Whey protein ingestion did not alter glycemia, but increased circulating glucagon (peak 4-fold basal), insulin (peak 6-fold basal), amino acids, and urea while also reducing free fatty acid (FFA) and glycerol concentrations. Interestingly, the postprandial insulin response was driven by both a stimulation of insulin secretion and marked reduction in hepatic insulin clearance. Whey protein ingestion resulted in a modest stimulation of EGP and Rd, both peaking at ∼20% above baseline 1 h after protein ingestion. These findings demonstrate that the ingestion of a single standard serving of whey protein can induce a euglycemic glucagon and insulin response that stimulates glucose flux. We speculate on a theory that could potentially explain how glucagon and insulin synergistically provide hardwired control of nitrogen and glucose homeostasis.NEW & NOTEWORTHY Protein ingestion concurrently stimulates glucagon and insulin secretion. Here we show that in normoglycemic males, ingestion of a single "standard" 25 g serving of rapidly absorbing whey protein drives a sufficiently large glucagon and insulin response, such that it simultaneously increases endogenous glucose production and glucose disposal. We speculate on a novel theory that could potentially explain how the antagonistic/synergistic actions of glucagon and insulin simultaneously provide tight control of glucose and nitrogen homeostasis.
Collapse
Affiliation(s)
- Teddy Ang
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Geelong, Victoria, Australia
- School of Exercise and Nutrition Science, Deakin University, Geelong, Victoria, Australia
| | - Shaun A Mason
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Geelong, Victoria, Australia
- School of Exercise and Nutrition Science, Deakin University, Geelong, Victoria, Australia
| | - Giang M Dao
- Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Clinton R Bruce
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Geelong, Victoria, Australia
- School of Exercise and Nutrition Science, Deakin University, Geelong, Victoria, Australia
| | - Greg M Kowalski
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Geelong, Victoria, Australia
- Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Victoria, Australia
| |
Collapse
|
18
|
Galderisi A, Carr ALJ, Martino M, Taylor P, Senior P, Dayan C. Quantifying beta cell function in the preclinical stages of type 1 diabetes. Diabetologia 2023; 66:2189-2199. [PMID: 37712956 PMCID: PMC10627950 DOI: 10.1007/s00125-023-06011-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 08/08/2023] [Indexed: 09/16/2023]
Abstract
Clinically symptomatic type 1 diabetes (stage 3 type 1 diabetes) is preceded by a pre-symptomatic phase, characterised by progressive loss of functional beta cell mass after the onset of islet autoimmunity, with (stage 2) or without (stage 1) measurable changes in glucose profile during an OGTT. Identifying metabolic tests that can longitudinally track changes in beta cell function is of pivotal importance to track disease progression and measure the effect of disease-modifying interventions. In this review we describe the metabolic changes that occur in the early pre-symptomatic stages of type 1 diabetes with respect to both insulin secretion and insulin sensitivity, as well as the measurable outcomes that can be derived from the available tests. We also discuss the use of metabolic modelling to identify insulin secretion and sensitivity, and the measurable changes during dynamic tests such as the OGTT. Finally, we review the role of risk indices and minimally invasive measures such as those derived from the use of continuous glucose monitoring.
Collapse
Affiliation(s)
| | - Alice L J Carr
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Mariangela Martino
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Peter Taylor
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Peter Senior
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Colin Dayan
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK.
| |
Collapse
|
19
|
Zaharia OP, Antoniou S, Bobrov P, Karusheva Y, Bódis K, Kupriyanova Y, Schrauwen-Hinderling V, Gastaldelli A, Szendroedi J, Wagner R, Burkart V, Roden M. Reduced Insulin Clearance Differently Relates to Increased Liver Lipid Content and Worse Glycemic Control in Recent-Onset Type 2 and Type 1 Diabetes. Diabetes Care 2023; 46:2232-2239. [PMID: 37874983 DOI: 10.2337/dc23-1267] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 09/15/2023] [Indexed: 10/26/2023]
Abstract
OBJECTIVE Diabetes may feature impaired insulin kinetics, which could be aggravated by altered hepatic metabolism and glycemic control. Thus, we examined insulin clearance and its possible determinants in individuals with recent-onset diabetes. RESEARCH DESIGN AND METHODS Participants of the German Diabetes Study (GDS) with type 1 diabetes (T1D) (n = 306), type 2 diabetes (T2D) (n = 489), or normal glucose tolerance (control [CON]) (n = 167) underwent hyperinsulinemic-euglycemic clamps for assessment of whole-body insulin sensitivity (M value) and insulin clearance (ICCLAMP). Insulin clearance rates were further calculated during intravenous glucose tolerance tests (ICIVGTT) and mixed-meal tests (ICMMT). Hepatocellular lipid content (HCL) was quantified with 1H-MRS. RESULTS Both T1D and T2D groups had lower ICCLAMP (0.12 ± 0.07 and 0.21 ± 0.06 vs. 0.28 ± 0.14 arbitrary units [a.u.], respectively, all P < 0.05) and ICMMT (0.71 ± 0.35 and 0.99 ± 0.33 vs. 1.20 ± 0.36 a.u., all P < 0.05) than CON. In T1D, ICCLAMP, ICIVGTT, and ICMMT correlated negatively with HbA1c (all P < 0.05). M value correlated positively with ICIVGTT in CON and T2D (r = 0.199 and r = 0.178, P < 0.05) and with ICMMT in CON (r = 0.176, P < 0.05). HCL negatively associated with ICIVGTT and ICMMT in T2D (r = -0.005 and r = -0.037) and CON (r = -0.127 and r = -0.058, all P < 0.05). In line, T2D or CON subjects with steatosis featured lower ICMMT than those without steatosis (both P < 0.05). CONCLUSIONS Insulin clearance is reduced in both T1D and T2D within the first year after diagnosis but correlates negatively with liver lipid content rather in T2D. Moreover, insulin clearance differently associates with glycemic control and insulin sensitivity in each diabetes type, which may suggest specific mechanisms affecting insulin kinetics.
Collapse
Affiliation(s)
- Oana-Patricia Zaharia
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital, Heinrich Heine University, Düsseldorf, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, Düsseldorf, Germany
| | - Sofia Antoniou
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
| | - Pavel Bobrov
- German Center for Diabetes Research, Partner Düsseldorf, Düsseldorf, Germany
- Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
| | - Yanislava Karusheva
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, Düsseldorf, Germany
| | - Kálmán Bódis
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital, Heinrich Heine University, Düsseldorf, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, Düsseldorf, Germany
| | - Yuliya Kupriyanova
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, Düsseldorf, Germany
| | - Vera Schrauwen-Hinderling
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, Düsseldorf, Germany
| | - Amalia Gastaldelli
- Institute of Clinical Physiology, Consiglio Nazionale delle Ricerche (CNR), Pisa, Italy
| | - Julia Szendroedi
- Department of Internal Medicine I and Clinical Chemistry, University Hospital Heidelberg, Heidelberg, Germany
| | - Robert Wagner
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital, Heinrich Heine University, Düsseldorf, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, Düsseldorf, Germany
| | - Volker Burkart
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, Düsseldorf, Germany
| | - Michael Roden
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital, Heinrich Heine University, Düsseldorf, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
20
|
Sugimoto H, Hironaka KI, Yamada T, Sakaguchi K, Ogawa W, Kuroda S. DI/cle, a Measure Consisting of Insulin Sensitivity, Secretion, and Clearance, Captures Diabetic States. J Clin Endocrinol Metab 2023; 108:3080-3089. [PMID: 37406246 PMCID: PMC10655546 DOI: 10.1210/clinem/dgad392] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 06/26/2023] [Accepted: 06/30/2023] [Indexed: 07/07/2023]
Abstract
CONTEXT Insulin clearance is implicated in regulation of glucose homeostasis independently of insulin sensitivity and insulin secretion. OBJECTIVE To understand the relation between blood glucose and insulin sensitivity, secretion, and clearance. METHODS We performed a hyperglycemic clamp, a hyperinsulinemic-euglycemic clamp, and an oral glucose tolerance test (OGTT) in 47, 16, and 49 subjects with normal glucose tolerance (NGT), impaired glucose tolerance (IGT), and type 2 diabetes mellitus (T2DM), respectively. Mathematical analyses were retrospectively performed on this dataset. RESULTS The disposition index (DI), defined as the product of insulin sensitivity and secretion, showed a weak correlation with blood glucose levels, especially in IGT (r = 0.04; 95% CI, -0.63 to 0.44). However, an equation relating DI, insulin clearance, and blood glucose levels was well conserved regardless of the extent of glucose intolerance. As a measure of the effect of insulin, we developed an index, designated disposition index/clearance, (DI/cle) that is based on this equation and corresponds to DI divided by the square of insulin clearance. DI/cle was not impaired in IGT compared with NGT, possibly as a result of a decrease in insulin clearance in response to a reduction in DI, whereas it was impaired in T2DM relative to IGT. Moreover, DI/cle estimated from a hyperinsulinemic-euglycemic clamp, OGTT, or a fasting blood test were significantly correlated with that estimated from 2 clamp tests (r = 0.52; 95% CI, 0.37 to 0.64, r = 0.43; 95% CI, 0.24 to 0.58, r = 0.54; 95% CI, 0.38 to 0.68, respectively). CONCLUSION DI/cle can serve as a new indicator for the trajectory of changes in glucose tolerance.
Collapse
Affiliation(s)
- Hikaru Sugimoto
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Ken-ichi Hironaka
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo 113-0033, Japan
| | - Tomoko Yamada
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo 650-0017, Japan
| | - Kazuhiko Sakaguchi
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo 650-0017, Japan
| | - Wataru Ogawa
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo 650-0017, Japan
| | - Shinya Kuroda
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo 113-0033, Japan
| |
Collapse
|
21
|
Franceschi R, Fintini D, Ravà L, Mariani M, Aureli A, Inzaghi E, Pedicelli S, Deodati A, Bizzarri C, Cappa M, Cianfarani S, Manco M. Insulin Clearance at the Pubertal Transition in Youth with Obesity and Steatosis Liver Disease. Int J Mol Sci 2023; 24:14963. [PMID: 37834412 PMCID: PMC10573227 DOI: 10.3390/ijms241914963] [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: 08/29/2023] [Revised: 09/29/2023] [Accepted: 10/02/2023] [Indexed: 10/15/2023] Open
Abstract
No data are available on insulin clearance (ClI) trends during the pubertal transition. The aim of this study was to investigate in 973 youths with obesity whether ClI in fasting and post-oral glucose challenge (OGTT) conditions varies at the pubertal transition in relation to the severity of obesity and the presence of steatosis liver disease (SLD). The severity of obesity was graded according to the Centers for Disease Control. SLD was graded as absent, mild and severe based on alanine amino transferase levels. ClI was defined as the molar ratio of fasting C-peptide to insulin and of the areas under the insulin to glucose curves during an OGTT. In total, 35% of participants were prepubertal, 72.6% had obesity class II, and 52.6% had mild SLD. Fasting ClI (nmol/pmol × 10-2) was significantly lower in pubertal [0.11 (0.08-0.14)] than in prepubertal individuals [0.12 (0.09-0.16)] and higher in class III [0.15 (0.11-0.16)] than in class I obesity [0.11 (0.09-0.14)]. OGTT ClI was higher in boys [0.08 (0.06-0.10)] than in girls [0.07 (0.06-0.09)]; in prepubertal [0.08 (0.06-0.11)] than in pubertal individuals [0.07 (0.05-0.09)]; in class III [0.14 (0.08-0.17)] than in class I obesity [0.07 (0.05-0.10)]; and in severe SLD [0.09 (0.04-0.14)] than in no steatosis [0.06 (0.04-0.17)]. It was lower in participants with prediabetes [0.06 (0.04-0.07)]. OGTT ClI was lower in youths with obesity at puberty along with insulin sensitivity and greater secretion. The findings suggest that the initial increase in ClI in youth with severe obesity and SLD is likely to compensate for hyperinsulinemia and its subsequent decrease at the onset of prediabetes and other metabolic abnormalities.
Collapse
Affiliation(s)
- Roberto Franceschi
- Pediatric Department, S. Chiara Hospital of Trento, APSS, 38121 Trento, Italy;
| | - Danilo Fintini
- Diabetes and Growth Disorders Unit, Bambino Gesù Children’s Hospital, IRCCS, 00168 Rome, Italy; (D.F.); (M.M.); (A.A.); (E.I.); (S.P.); (A.D.); (C.B.); or (S.C.)
| | - Lucilla Ravà
- Clinical Epidemiology, Bambino Gesù Children’s Hospital, IRCCS, 00168 Rome, Italy
| | - Michela Mariani
- Diabetes and Growth Disorders Unit, Bambino Gesù Children’s Hospital, IRCCS, 00168 Rome, Italy; (D.F.); (M.M.); (A.A.); (E.I.); (S.P.); (A.D.); (C.B.); or (S.C.)
| | - Alessia Aureli
- Diabetes and Growth Disorders Unit, Bambino Gesù Children’s Hospital, IRCCS, 00168 Rome, Italy; (D.F.); (M.M.); (A.A.); (E.I.); (S.P.); (A.D.); (C.B.); or (S.C.)
| | - Elena Inzaghi
- Diabetes and Growth Disorders Unit, Bambino Gesù Children’s Hospital, IRCCS, 00168 Rome, Italy; (D.F.); (M.M.); (A.A.); (E.I.); (S.P.); (A.D.); (C.B.); or (S.C.)
| | - Stefania Pedicelli
- Diabetes and Growth Disorders Unit, Bambino Gesù Children’s Hospital, IRCCS, 00168 Rome, Italy; (D.F.); (M.M.); (A.A.); (E.I.); (S.P.); (A.D.); (C.B.); or (S.C.)
| | - Annalisa Deodati
- Diabetes and Growth Disorders Unit, Bambino Gesù Children’s Hospital, IRCCS, 00168 Rome, Italy; (D.F.); (M.M.); (A.A.); (E.I.); (S.P.); (A.D.); (C.B.); or (S.C.)
| | - Carla Bizzarri
- Diabetes and Growth Disorders Unit, Bambino Gesù Children’s Hospital, IRCCS, 00168 Rome, Italy; (D.F.); (M.M.); (A.A.); (E.I.); (S.P.); (A.D.); (C.B.); or (S.C.)
| | - Marco Cappa
- Research Unit, Innovative Therapies for Endocrinopathies, Scientific Directorate, Bambino Gesù Children’s Hospital, IRCCS, 00168 Rome, Italy;
| | - Stefano Cianfarani
- Diabetes and Growth Disorders Unit, Bambino Gesù Children’s Hospital, IRCCS, 00168 Rome, Italy; (D.F.); (M.M.); (A.A.); (E.I.); (S.P.); (A.D.); (C.B.); or (S.C.)
- Department of Systems Medicine, University of Rome ‘Tor Vergata’, 00168 Rome, Italy
- Department of Women’s and Children’s Health, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Melania Manco
- Research Unit of Predictive and Preventive Medicine, Bambino Gesù Children’s Hospital, 00146 Rome, Italy
| |
Collapse
|
22
|
Finn E, Severn C, Pyle L, Garrish J, Vigers T, Behn CGD, Zeitler PS, Sagel SD, Nadeau KJ, Chan CL. Hypoglycemia in children and young adults with cystic fibrosis during oral glucose tolerance testing vs. continuous glucose monitoring. Pediatr Pulmonol 2023; 58:2495-2504. [PMID: 37350354 PMCID: PMC10538856 DOI: 10.1002/ppul.26533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 04/27/2023] [Accepted: 05/27/2023] [Indexed: 06/24/2023]
Abstract
BACKGROUND Hypoglycemia is common in people with cystic fibrosis (pwCF) during oral glucose tolerance tests (OGTTs) and in the free-living setting, yet its pathophysiology remains unclear. OBJECTIVE To evaluate hypoglycemia in children and young adults with CF by OGTT and continuous glucose monitoring (CGM). METHODS A 3-h OGTT was performed in children and young adults with CF and healthy controls (HC). Individuals were classified as experiencing hypoglycemia on OGTT (glucose <70 mg/dL) or not. Insulin, C-peptide, glucose, glucagon, and incretins were measured. CGM was performed for 7 days in the free-living setting. Measures of insulin sensitivity, beta cell function accounting for insulin sensitivity, and insulin clearance were calculated. RESULTS A total of 57 participants (40 CF and 17 HC) underwent assessment. Rates of hypoglycemia by OGTT were similar in pwCF (53%, 21/40) compared to HC (35%, 6/17), p = 0.23. PwCF compared to HC had higher A1c; on OGTT higher and later glucose peaks, later insulin peaks; and on CGM more glucose variability. CF Hypo+ versus CF Hypo- had higher lung function, higher insulin sensitivity, higher beta cell function accounting for insulin sensitivity, and decreased CGM variability. When comparing CF Hypo+ to HC Hypo+, although rates of hypoglycemia are similar, pwCF had blunted glucagon responses to hypoglycemia. OGTT hypoglycemia was not associated with CGM hypoglycemia in any group. CONCLUSION Youth with CF have increased insulin sensitivity and impaired glucagon response to hypoglycemia on OGTT. Hypoglycemia on OGTT did not associate with free-living hypoglycemia.
Collapse
Affiliation(s)
- Erin Finn
- Section of Pediatric Endocrinology, Department of Pediatrics, Children’s Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Cameron Severn
- Department of Biostatistics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Laura Pyle
- Department of Biostatistics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Justin Garrish
- Department of Applied Mathematics and Statistics, Colorado School of Mines, Golden, CO 80401, USA
| | - Timothy Vigers
- Section of Pediatric Endocrinology, Department of Pediatrics, Children’s Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Cecilia G. Diniz Behn
- Section of Pediatric Endocrinology, Department of Pediatrics, Children’s Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Applied Mathematics and Statistics, Colorado School of Mines, Golden, CO 80401, USA
| | - Philip S. Zeitler
- Section of Pediatric Endocrinology, Department of Pediatrics, Children’s Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Scott D. Sagel
- Section of Pediatric Pulmonology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Kristen J. Nadeau
- Section of Pediatric Endocrinology, Department of Pediatrics, Children’s Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Christine L. Chan
- Section of Pediatric Endocrinology, Department of Pediatrics, Children’s Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| |
Collapse
|
23
|
Galderisi A, Evans-Molina C, Martino M, Caprio S, Cobelli C, Moran A. β-Cell Function and Insulin Sensitivity in Youth With Early Type 1 Diabetes From a 2-Hour 7-Sample OGTT. J Clin Endocrinol Metab 2023; 108:1376-1386. [PMID: 36546354 PMCID: PMC10188312 DOI: 10.1210/clinem/dgac740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 12/15/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022]
Abstract
CONTEXT The oral minimal model is a widely accepted noninvasive tool to quantify both β-cell responsiveness and insulin sensitivity (SI) from glucose, C-peptide, and insulin concentrations during a 3-hour 9-point oral glucose tolerance test (OGTT). OBJECTIVE Here, we aimed to validate a 2-hour 7-point protocol against the 3-hour OGTT and to test how variation in early sampling frequency impacts estimates of β-cell responsiveness and SI. METHODS We conducted a secondary analysis on 15 lean youth with stage 1 type 1 diabetes (T1D; ≥ 2 islet autoantibodies with no dysglycemia) who underwent a 3-hour 9-point OGTT. The oral minimal model was used to quantitate β-cell responsiveness (φtotal) and insulin sensitivity (SI), allowing assessment of β-cell function by the disposition index (DI = φtotal × SI). Seven- and 5-point 2-hour OGTT protocols were tested against the 3-hour 9-point gold standard to determine agreement between estimates of φtotal and its dynamic and static components, SI, and DI across different sampling strategies. RESULTS The 2-hour estimates for the disposition index exhibited a strong correlation with 3-hour measures (r = 0.975; P < .001) with similar results for β-cell responsiveness and SI (r = 0.997 and r = 0.982; P < .001, respectively). The agreement of the 3 estimates between the 7-point 2-hour and 9-point 3-hour protocols fell within the 95% CI on the Bland-Altman grid with a median difference of 16.9% (-35.3 to 32.5), 0.2% (-0.6 to 1.3), and 14.9% (-1.4 to 28.3) for DI, φtotal, and SI. Conversely, the 5-point protocol did not provide reliable estimates of φ dynamic and static components. CONCLUSION The 2-hour 7-point OGTT is reliable in individuals with stage 1 T1D for assessment of β-cell responsiveness, SI, and DI. Incorporation of these analyses into current 2-hour diabetes staging and monitoring OGTTs offers the potential to more accurately quantify risk of progression in the early stages of T1D.
Collapse
Affiliation(s)
- Alfonso Galderisi
- Department of Woman and Child's Health, University of Padova,
35128 Padua, Italy
| | - Carmella Evans-Molina
- Center for Diabetes and Metabolic Diseases, Indiana
University, Indianapolis, Indiana 46202, USA
| | - Mariangela Martino
- Department of Woman and Child's Health, University of Padova,
35128 Padua, Italy
| | - Sonia Caprio
- Department of Pediatrics, Yale University, New
Haven, Connecticut 06520, USA
| | - Claudio Cobelli
- Department of Woman and Child's Health, University of Padova,
35128 Padua, Italy
| | - Antoinette Moran
- Department of Pediatrics, University of Minnesota,
Minneapolis, Minnesota 55454, USA
| |
Collapse
|
24
|
Meneses MJ, Patarrão RS, Pinheiro T, Coelho I, Carriço N, Marques AC, Romão A, Nabais J, Fortunato E, Raposo JF, Macedo MP. Leveraging the future of diagnosis and management of diabetes: From old indexes to new technologies. Eur J Clin Invest 2023; 53:e13934. [PMID: 36479853 DOI: 10.1111/eci.13934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/15/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Diabetes is a heterogeneous and multifactorial disease. However, glycemia and glycated hemoglobin have been the focus of diabetes diagnosis and management for the last decades. As diabetes management goes far beyond glucose control, it has become clear that assessment of other biochemical parameters gives a much wider view of the metabolic state of each individual, enabling a precision medicine approach. METHODS In this review, we summarize and discuss indexes that have been used in epidemiological studies and in the clinical practice. RESULTS Indexes of insulin secretion, sensitivity/resistance and metabolism have been developed and validated over the years to account also with insulin, C-peptide, triglycerides or even anthropometric measures. Nevertheless, each one has their own objective and consequently, advantages and disadvantages for specific cases. Thus, we discuss how new technologies, namely new sensors but also new softwares/applications, can improve the diagnosis and management of diabetes, both for healthcare professionals but also for caretakers and, importantly, to promote the empowerment of people living with diabetes. CONCLUSIONS In long-term, the solution for a better diabetes management would be a platform that allows to integrate all sorts of relevant information for the person with diabetes and for the healthcare practitioners, namely glucose, insulin and C-peptide or, in case of need, other parameters/indexes at home, sometimes more than once a day. This solution would allow a better and simpler disease management, more adequate therapeutics thereby improving patients' quality of life and reducing associated costs.
Collapse
Affiliation(s)
- Maria João Meneses
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisbon, Portugal.,DECSIS II Iberia, Évora, Portugal
| | - Rita Susana Patarrão
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Tomás Pinheiro
- CENIMAT i3N, Materials Science Department, Faculty of Science and Technology, Universidade NOVA de Lisboa and CEMOP/UNINOVA, Caparica, Portugal
| | - Inês Coelho
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisbon, Portugal
| | | | - Ana Carolina Marques
- CENIMAT i3N, Materials Science Department, Faculty of Science and Technology, Universidade NOVA de Lisboa and CEMOP/UNINOVA, Caparica, Portugal
| | | | - João Nabais
- Comprehensive Health Research Centre (CHRC), Departamento de Ciências Médicas e da Saúde, Escola de Saúde e Desenvolvimento Humano, Universidade de Évora, Évora, Portugal
| | - Elvira Fortunato
- CENIMAT i3N, Materials Science Department, Faculty of Science and Technology, Universidade NOVA de Lisboa and CEMOP/UNINOVA, Caparica, Portugal
| | - João Filipe Raposo
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisbon, Portugal.,APDP - Diabetes Portugal - Education and Research Center, Lisbon, Portugal
| | - Maria Paula Macedo
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisbon, Portugal.,APDP - Diabetes Portugal - Education and Research Center, Lisbon, Portugal
| |
Collapse
|
25
|
Abstract
Insulin action is impaired in type 2 diabetes. The functions of the hormone are an integrated product of insulin secretion from pancreatic β-cells and insulin clearance by receptor-mediated endocytosis and degradation, mostly in liver (hepatocytes) and, to a lower extent, in extrahepatic peripheral tissues. Substantial evidence indicates that genetic or acquired abnormalities of insulin secretion or action predispose to type 2 diabetes. In recent years, along with the discovery of the molecular foundation of receptor-mediated insulin clearance, such as through the membrane glycoprotein CEACAM1, a consensus has begun to emerge that reduction of insulin clearance contributes to the disease process. In this review, we consider the evidence suggesting a pathogenic role for reduced insulin clearance in insulin resistance, obesity, hepatic steatosis, and type 2 diabetes.
Collapse
Affiliation(s)
- Sonia M Najjar
- Department of Biomedical Sciences and the Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA;
| | - Sonia Caprio
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Amalia Gastaldelli
- Cardiometabolic Risk Unit, Institute of Clinical Physiology-National Research Council, Pisa, Italy
| |
Collapse
|
26
|
Sharma A, Lee-Ødegård S, Qvigstad E, Sommer C, Sattar N, Gill JMR, Gulseth HL, Sollid ST, Nermoen I, Birkeland KI. β-Cell Function, Hepatic Insulin Clearance, and Insulin Sensitivity in South Asian and Nordic Women After Gestational Diabetes Mellitus. Diabetes 2022; 71:2530-2538. [PMID: 36112815 DOI: 10.2337/db22-0622] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 09/14/2022] [Indexed: 01/11/2023]
Abstract
South Asian women have a higher risk of type 2 diabetes after gestational diabetes mellitus (GDM) than Nordic women; however, the mechanisms behind this difference remain unclear. We investigated insulin sensitivity, β-cell function, and hepatic insulin clearance in 179 South Asian and 108 Nordic women ∼17 months after GDM (mean age 35.3 years, BMI 29.1 kg/m2) by oral glucose tolerance test using deconvolution of C-peptide kinetics. Thirty-one percent of South Asian and 53% of Nordic participants were normoglycemic at the time of measurement. South Asian women had higher areas under the curve (AUCs) for glucose, prehepatic insulin, and peripheral insulin and lower insulin sensitivity, disposition index, and fasting hepatic insulin clearance than Nordic women. In the group with prediabetes or diabetes, South Asian women had similar AUCs for glucose and prehepatic insulin but a higher AUC for peripheral insulin, lower disposition index, and lower fasting hepatic insulin clearance than Nordic women. The waist-to-height ratio mediated ∼25-40% of the ethnic differences in insulin sensitivity in participants with normoglycemia. Overall, our novel data revealed that South Asian women with normoglycemia after GDM showed lower insulin secretion for a given insulin resistance and lower hepatic insulin clearance than Nordic women. South Asian women are at high risk of developing type 2 diabetes after GDM, and preventive efforts should be prioritized.
Collapse
Affiliation(s)
- Archana Sharma
- Department of Endocrinology, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Elisabeth Qvigstad
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
| | - Christine Sommer
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, U.K
| | - Jason M R Gill
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, U.K
| | | | - Stina T Sollid
- Department of Medicine, Drammen Hospital, Vestre Viken Health Trust, Drammen, Norway
| | - Ingrid Nermoen
- Department of Endocrinology, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Kåre I Birkeland
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
27
|
Subramanian V, Bagger JI, Holst JJ, Knop FK, Vilsbøll T. A glucose-insulin-glucagon coupled model of the isoglycemic intravenous glucose infusion experiment. Front Physiol 2022; 13:911616. [PMID: 36148302 PMCID: PMC9485803 DOI: 10.3389/fphys.2022.911616] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
Type 2 diabetes (T2D) is a pathophysiology that is characterized by insulin resistance, beta- and alpha-cell dysfunction. Mathematical models of various glucose challenge experiments have been developed to quantify the contribution of insulin and beta-cell dysfunction to the pathophysiology of T2D. There is a need for effective extended models that also capture the impact of alpha-cell dysregulation on T2D. In this paper a delay differential equation-based model is developed to describe the coupled glucose-insulin-glucagon dynamics in the isoglycemic intravenous glucose infusion (IIGI) experiment. As the glucose profile in IIGI is tailored to match that of a corresponding oral glucose tolerance test (OGTT), it provides a perfect method for studying hormone responses that are in the normal physiological domain and without the confounding effect of incretins and other gut mediated factors. The model was fit to IIGI data from individuals with and without T2D. Parameters related to glucagon action, suppression, and secretion as well as measures of insulin sensitivity, and glucose stimulated response were determined simultaneously. Significant impairment in glucose dependent glucagon suppression was observed in patients with T2D (duration of T2D: 8 (6-36) months) relative to weight matched control subjects (CS) without diabetes (k1 (mM)-1: 0.16 ± 0.015 (T2D, n = 7); 0.26 ± 0.047 (CS, n = 7)). Insulin action was significantly lower in patients with T2D (a1 (10 pM min)-1: 0.000084 ± 0.0000075 (T2D); 0.00052 ± 0.00015 (CS)) and the Hill coefficient in the equation for glucose dependent insulin response was found to be significantly different in T2D patients relative to CS (h: 1.4 ± 0.15; 1.9 ± 0.14). Trends in parameters with respect to fasting plasma glucose, HbA1c and 2-h glucose values are also presented. Significantly, a negative linear relationship is observed between the glucagon suppression parameter, k1, and the three markers for diabetes and is thus indicative of the role of glucagon in exacerbating the pathophysiology of diabetes (Spearman Rank Correlation: (n = 12; (-0.79, 0.002), (-0.73,.007), (-0.86,.0003)) respectively).
Collapse
Affiliation(s)
- Vijaya Subramanian
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Jonatan I. Bagger
- Center for Clinical Metabolic Research, Herlev and Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Clinical Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Jens J. Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Filip K. Knop
- Center for Clinical Metabolic Research, Herlev and Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Clinical Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tina Vilsbøll
- Center for Clinical Metabolic Research, Herlev and Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Clinical Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
28
|
Åkerström T, Stolpe MN, Widmer R, Dejgaard TF, Højberg JM, Møller K, Hansen JS, Trinh B, Holst JJ, Thomsen C, Pedersen BK, Ellingsgaard H. Endurance Training Improves GLP-1 Sensitivity and Glucose Tolerance in Overweight Women. J Endocr Soc 2022; 6:bvac111. [PMID: 35935071 PMCID: PMC9351379 DOI: 10.1210/jendso/bvac111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Indexed: 11/19/2022] Open
Abstract
Context and objective Obesity and inactivity are risk factors for developing impaired glucose tolerance characterized by insulin resistance and reduced beta-cell function. The stimulatory effect of glucagon-like peptide 1 (GLP-1) on insulin secretion is also impaired in obese, inactive individuals. The aim of this study was to investigate whether endurance training influences beta-cell sensitivity to GLP-1. Participants and intervention Twenty-four female participants, age 46 ± 2 years, body mass index 32.4 ± 0.9 kg/m2, and maximal oxygen consumption 24.7 ± 0.8 mL/kg/min participated in a 10-week exercise training study. Methods Beta-cell sensitivity to GLP-1 was assessed in a subset of participants (n = 6) during a 120-minute hyperglycemic glucose clamp (8.5 mM) including a 1-hour GLP-1 (7-36 amide) infusion (0.4 pmol/kg/min). Changes in glucose tolerance, body composition, and cardiorespiratory fitness were assessed by oral glucose tolerance tests (OGTTs), dual-energy X-ray absorptiometry scans, magnetic resonance scans, and maximal oxygen consumption (VO2max) tests, respectively. Results The c-peptide response to infusion of GLP-1 increased 28 ± 3% (P < 0.05) toward the end of the hyperglycemic clamp. The insulin response remained unchanged. Training improved glucose tolerance and reduced GLP-1, insulin, and glucagon levels during the OGTTs. Training increased VO2max (from 24.7 ± 0.8 to 27.0 ± 0.7 mL/kg/min; P < 0.05) and reduced visceral fat volume (from 4176 ± 265 to 3888 ± 266 cm3; P < 0.01). Conclusion Along with improved glycemic control, endurance training improved beta-cell sensitivity to GLP-1 in overweight women. The study was deemed not to constitute a clinical trial and was not registered as such.
Collapse
Affiliation(s)
- Thorbjörn Åkerström
- Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Rigshospitalet, Copenhagen University Hospital – Rigshospitalet , DK 2100 Copenhagen , Denmark
- Diabetes Pharmacology 1, Novo Nordisk A/S , Maaløv , Denmark
| | - Malene N Stolpe
- Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Rigshospitalet, Copenhagen University Hospital – Rigshospitalet , DK 2100 Copenhagen , Denmark
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen , DK 2200 Copenhagen , Denmark
| | - Renate Widmer
- Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Rigshospitalet, Copenhagen University Hospital – Rigshospitalet , DK 2100 Copenhagen , Denmark
| | - Thomas F Dejgaard
- Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Rigshospitalet, Copenhagen University Hospital – Rigshospitalet , DK 2100 Copenhagen , Denmark
| | - Jens M Højberg
- Department of Cardiothoracic Anesthesiology and Intensive Care, Rigshospitalet , DK 2100 Copenhagen , Denmark
| | - Kirsten Møller
- Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Rigshospitalet, Copenhagen University Hospital – Rigshospitalet , DK 2100 Copenhagen , Denmark
- Intensive Care Unit 4131, Rigshospitalet , DK 2100 Copenhagen , Denmark
| | - Jakob S Hansen
- Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Rigshospitalet, Copenhagen University Hospital – Rigshospitalet , DK 2100 Copenhagen , Denmark
- Novo Nordisk A/S , Søborg , Denmark
| | - Beckey Trinh
- Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Rigshospitalet, Copenhagen University Hospital – Rigshospitalet , DK 2100 Copenhagen , Denmark
| | - Jens J Holst
- Department of Biomedical Sciences and the NNF Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen , DK 2200 Copenhagen , Denmark
| | - Carsten Thomsen
- Department of Radiology, Rigshospitalet, University of Copenhagen , DK 2100 Copenhagen , Denmark
| | - Bente K Pedersen
- Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Rigshospitalet, Copenhagen University Hospital – Rigshospitalet , DK 2100 Copenhagen , Denmark
| | - Helga Ellingsgaard
- Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Rigshospitalet, Copenhagen University Hospital – Rigshospitalet , DK 2100 Copenhagen , Denmark
| |
Collapse
|
29
|
Tricò D, Mengozzi A, Baldi S, Bizzotto R, Olaniru O, Toczyska K, Huang GC, Seghieri M, Frascerra S, Amiel SA, Persaud S, Jones P, Mari A, Natali A. Lipid-induced glucose intolerance is driven by impaired glucose kinetics and insulin metabolism in healthy individuals. Metabolism 2022; 134:155247. [PMID: 35760117 DOI: 10.1016/j.metabol.2022.155247] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 11/19/2022]
Abstract
AIMS Hypertriglyceridemia is associated with an increased risk of type 2 diabetes. We aimed to comprehensively examine the effects of hypertriglyceridemia on major glucose homeostatic mechanisms involved in diabetes progression. METHODS In this randomized, cross-over, single-blinded study, two dual-labeled, 3-hour oral glucose tolerance tests were performed during 5-hour intravenous infusions of either 20 % Intralipid or saline in 12 healthy subjects (age 27.9 ± 2.6 years, 11 men, BMI 22.6 ± 1.4 kg/m2) to evaluate lipid-induced changes in insulin metabolism and glucose kinetics. Insulin sensitivity, β cell secretory function, and insulin clearance were assessed by modeling glucose, insulin and C-peptide data. Intestinal glucose absorption, endogenous glucose production, and glucose clearance were assessed from glucose tracers. The effect of triglycerides on β-cell secretory function was examined in perifusion experiments in murine pseudoislets and human pancreatic islets. RESULTS Mild acute hypertriglyceridemia impaired oral glucose tolerance (mean glucose: +0.9 [0.3, 1.5] mmol/L, p = 0.008) and whole-body insulin sensitivity (Matsuda index: -1.67 [-0.50, -2.84], p = 0.009). Post-glucose hyperinsulinemia (mean insulin: +99 [17, 182] pmol/L, p = 0.009) resulted from reduced insulin clearance (-0.16 [-0.32, -0.01] L min-1 m-2, p = 0.04) and enhanced hyperglycemia-induced total insulin secretion (+11.9 [1.1, 22.8] nmol/m2, p = 0.02), which occurred despite a decline in model-derived β cell glucose sensitivity (-41 [-74, -7] pmol min-1 m-2 mmol-1 L, p = 0.04). The analysis of tracer-derived glucose metabolic fluxes during lipid infusion revealed lower glucose clearance (-96 [-152, -41] mL/kgFFM, p = 0.005), increased 2-hour oral glucose absorption (+380 [42, 718] μmol/kgFFM, p = 0.04) and suppressed endogenous glucose production (-448 [-573, -123] μmol/kgFFM, p = 0.005). High-physiologic triglyceride levels increased acute basal insulin secretion in murine pseudoislets (+11 [3, 19] pg/aliquot, p = 0.02) and human pancreatic islets (+286 [59, 512] pg/islet, p = 0.02). CONCLUSION Our findings support a critical role for hypertriglyceridemia in the pathogenesis of type 2 diabetes in otherwise healthy individuals and dissect the glucose homeostatic mechanisms involved, encompassing insulin sensitivity, β cell function and oral glucose absorption.
Collapse
Affiliation(s)
- Domenico Tricò
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.
| | - Alessandro Mengozzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy; Institute of Life Sciences, Sant'Anna School of Advanced Studies, Pisa, Italy
| | - Simona Baldi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Roberto Bizzotto
- Institute of Neuroscience, National Research Council, Padua, Italy
| | - Oladapo Olaniru
- Department of Diabetes, School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Klaudia Toczyska
- Department of Diabetes, School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Guo Cai Huang
- Department of Diabetes, School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Marta Seghieri
- Diabetes and Metabolic Diseases Unit, "San Giovanni Di Dio" Hospital, Florence, Italy
| | - Silvia Frascerra
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Stephanie A Amiel
- Department of Diabetes, School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Shanta Persaud
- Department of Diabetes, School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Peter Jones
- Department of Diabetes, School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Andrea Mari
- Institute of Neuroscience, National Research Council, Padua, Italy
| | - Andrea Natali
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
30
|
Abdul-Ghani T, Puckett C, Migahid O, Abdelgani S, Migahed A, Adams J, Triplitt C, DeFronzo R, Jayyousi A, Abdul-Ghani M. Type 2 diabetes subgroups and response to glucose-lowering therapy: Results from the EDICT and Qatar studies. Diabetes Obes Metab 2022; 24:1810-1818. [PMID: 35581905 DOI: 10.1111/dom.14767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/20/2022] [Accepted: 05/08/2022] [Indexed: 11/29/2022]
Abstract
AIM To examine the efficacy of glucose-lowering medications in subgroups of patients with type 2 diabetes mellitus (T2DM). RESEARCH DESIGN AND METHODS Cluster analysis was performed in participants in the Efficacy and Durability of Initial Combination Therapy for Type 2 Diabetes (EDICT) study and the Qatar study using age, body mass index (BMI), glycated haemoglobin (HbA1c), and homeostatic model assessment of insulin resistance (HOMA-IR) and beta-cell function (HOMA-β). Participants also underwent an oral glucose tolerance test with measurement of plasma glucose, insulin and C-peptide concentrations to derive independent measures of insulin secretion and insulin sensitivity. The response to glucose-lowering therapies (change in HbA1c) was measured in each participant cluster for 3 years. RESULTS Three distinct and comparable clusters/groups of T2DM patients were identified in both the EDICT and Qatar studies. Participants in Group 1 had the highest HbA1c and manifested severe insulin deficiency. Participants in Group 3 had comparable insulin sensitivity to those in Group 1 but better beta-cell function and better glucose control. Participants in Group 2 had the highest BMI with severe insulin resistance accompanied by marked hyperinsulinaemia, which was primarily attributable to decreased insulin clearance. Unexpectedly, participants in Group 1 had better response to combination therapy with pioglitazone plus exenatide than with insulin therapy or metformin sequentially followed by glipizide and basal insulin, while participants in Group 2 responded equally well to both therapies despite very severe insulin resistance. CONCLUSION Distinct metabolic phenotypes characterize different T2DM clusters and differential responses to glucose-lowering therapies. Participants with severe insulin deficiency respond better to agents that preserve beta-cell function, while, surprisingly, patients with severe insulin resistance did not respond favourably to insulin sensitizers.
Collapse
Affiliation(s)
- Tamam Abdul-Ghani
- Division of Diabetes, University of Texas Health Science Center and Texas Diabetes Institute, San Antonio, Texas, USA
| | - Curtiss Puckett
- Division of Diabetes, University of Texas Health Science Center and Texas Diabetes Institute, San Antonio, Texas, USA
| | | | - Siham Abdelgani
- Division of Diabetes, University of Texas Health Science Center and Texas Diabetes Institute, San Antonio, Texas, USA
| | | | - John Adams
- Division of Diabetes, University of Texas Health Science Center and Texas Diabetes Institute, San Antonio, Texas, USA
| | - Curtis Triplitt
- Division of Diabetes, University of Texas Health Science Center and Texas Diabetes Institute, San Antonio, Texas, USA
| | - Ralph DeFronzo
- Division of Diabetes, University of Texas Health Science Center and Texas Diabetes Institute, San Antonio, Texas, USA
| | | | - Muhammad Abdul-Ghani
- Division of Diabetes, University of Texas Health Science Center and Texas Diabetes Institute, San Antonio, Texas, USA
| |
Collapse
|
31
|
Beaudry KM, Surdi JC, Mari A, Devries MC. Exercise mode influences post-exercise glucose sensitivity and insulin clearance in young, healthy males and females in a sex-dependent manner: A randomized control trial. Physiol Rep 2022; 10:e15354. [PMID: 35785485 PMCID: PMC9251832 DOI: 10.14814/phy2.15354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/11/2022] [Accepted: 05/21/2022] [Indexed: 06/15/2023] Open
Abstract
Type 2 diabetes (T2D) risk is lower in females than males. It has been reported that females have greater pancreatic 𝛽-cell function than males, which may at least in part contribute to the T2D risk in females. 𝛽-cell function is influenced by exercise training; however, previous trials comparing 𝛽-cell function between the sexes have not included participants matched for training status. Furthermore, the acute effects of different modes of exercise on 𝛽-cell function, and whether sex inherently influences these effects, are largely unexamined. Males and females (12/sex) completed a 120-min oral glucose tolerance test (OGTT) at rest (CON) and following acute bouts of high-intensity interval exercise (HIIE), moderate intensity continuous (MIC) exercise, and low-load high-repetition (LLHR) resistance exercise to assess whether sex inherently influences baseline and/or post-exercise pancreatic function in the absence of pathology. We found no sex differences in basal pancreatic 𝛽-cell function. Females had greater basal insulin clearance following MIC exercise compared to males (p = 0.01) and males tended to have a higher potentiation ratio following HIIE (p = 0.07). Females also had lower glucose sensitivity following MIC exercise compared to HIIE (p = 0.007) and LLHR (p = 0.003). Insulin clearance during the OGTT was greater following HIIE as compared with CON and MIC exercise (p = 0.02). 2-H oral glucose insulin sensitivity was greater following LLHR compared to CON (p = 0.01). Acute bouts of different modes of exercise do not differentially influence 𝛽-cell function but do influence insulin clearance and insulin sensitivity. Therefore, sex and exercise mode interact to differentially influence insulin clearance and glucose sensitivity.
Collapse
Affiliation(s)
| | - Julian C. Surdi
- Department of KinesiologyUniversity of WaterlooWaterlooCanada
| | - Andrea Mari
- Institute of Neuroscience, National Research CouncilPadovaItaly
| | | |
Collapse
|
32
|
Sugiyama S, Jinnouchi H, Hieshima K, Kurinami N, Jinnouchi K, Yoshida A, Suzuki T, Kajiwara K, Miyamoto F, Jinnouchi T. Potential Identification of Type 2 Diabetes with Elevated Insulin Clearance. NEJM EVIDENCE 2022; 1:EVIDoa2100052. [PMID: 38319210 DOI: 10.1056/evidoa2100052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
BACKGROUND: Decreased blood insulin concentrations resulting from reduced pancreatic β-cell insulin secretion and elevated insulin clearance (IC) could be involved in impaired glucose metabolism in diabetes. Recently, we reported a patient with type 2 diabetes mellitus (T2DM) who had decreased blood insulin concentrations and elevated IC. METHODS: For this study, we recruited patients with newly diagnosed, treatment-naïve T2DM and measured the metabolic clearance rate of insulin (MCRI) determined by a hyperinsulinemic-euglycemic clamp examination. We defined elevated IC as an MCRI of more than 700 ml/min/m2. Using this tentative cutoff, we identified patients with T2DM with elevated IC and investigated their clinical characteristics. RESULTS: We enrolled 101 patients in this study; 78.2% were men. Patients had a mean age of 54.1 years, a median body-mass index (BMI) of 25.1 kg/m2 (interquartile range [IQR], 22.9 to 28.4 kg/m2), a median hemoglobin A1c of 10.0% (IQR, 8.0 to 12.3%), and a median MCRI of 655 ml/min/m2 (IQR, 562 to 810 ml/min/m2). Our case definition for elevated IC was met by 44 patients whose median MCRI was 842 ml/min/m2 (IQR, 747 to 975 ml/min/m2) compared with those without elevated IC (570 ml/min/m2; IQR, 500 to 628 ml/min/m2). On the basis of this division, fasting blood glucose and insulin levels were 178 mg/dl (IQR, 140 to 218 mg/dl) and 4.2 mU/l (IQR, 2.7 to 5.5 mU/l), respectively, in patients with elevated IC compared with 146 mg/dl (IQR, 128 to 188 mg/dl) and 9.6 mU/l (IQR, 6.6 to 14.9 mU/l), respectively, in patients without elevated IC. The BMI of patients with elevated IC was 22.9 kg/m2 (IQR, 20.7 to 24.2 kg/m2) compared with 27.3 kg/m2 (IQR, 25.2 to 29.4 kg/m2) in patients who did not have elevated IC. There were no clinically significant differences in renal or hepatic function test results. CONCLUSIONS: Our data suggest that there is a group of patients with T2DM with elevated IC, and that they are nonobese and have decreased blood insulin concentrations. If confirmed, this novel form of T2DM could affect the treatment of such patients. (UMIN Clinical Trials Registry number, UMIN000032014.)
Collapse
Affiliation(s)
- Seigo Sugiyama
- Diabetes Care Center, Jinnouchi Hospital, Kumamoto, Japan
- Division of Cardiovascular Medicine, Diabetes Care Center, Jinnouchi Hospital, Kumamoto, Japan
| | - Hideaki Jinnouchi
- Diabetes Care Center, Jinnouchi Hospital, Kumamoto, Japan
- Division of Cardiovascular Medicine, Diabetes Care Center, Jinnouchi Hospital, Kumamoto, Japan
- Division of Preventive Cardiology, Department of Cardiovascular Medicine, Kumamoto University Hospital, Kumamoto, Japan
| | - Kunio Hieshima
- Diabetes Care Center, Jinnouchi Hospital, Kumamoto, Japan
- Infectious Disease Division, Diabetes Care Center, Jinnouchi Hospital, Kumamoto, Japan
| | - Noboru Kurinami
- Diabetes Care Center, Jinnouchi Hospital, Kumamoto, Japan
- Obesity Treatment Division, Diabetes Care Center, Jinnouchi Hospital, Kumamoto, Japan
| | - Katsunori Jinnouchi
- Diabetes Care Center, Jinnouchi Hospital, Kumamoto, Japan
- Division of Gastroenterology and Nephrology, Diabetes Care Center, Jinnouchi Hospital, Kumamoto, Japan
| | - Akira Yoshida
- Diabetes Care Center, Jinnouchi Hospital, Kumamoto, Japan
- Pharmacology Division, Diabetes Care Center, Jinnouchi Hospital, Kumamoto, Japan
| | - Tomoko Suzuki
- Diabetes Care Center, Jinnouchi Hospital, Kumamoto, Japan
- Division of Cardiovascular Medicine, Diabetes Care Center, Jinnouchi Hospital, Kumamoto, Japan
| | - Keizo Kajiwara
- Diabetes Care Center, Jinnouchi Hospital, Kumamoto, Japan
- Division of Cardiovascular Medicine, Diabetes Care Center, Jinnouchi Hospital, Kumamoto, Japan
| | - Fumio Miyamoto
- Diabetes Care Center, Jinnouchi Hospital, Kumamoto, Japan
- Ophthalmology Division, Diabetes Care Center, Jinnouchi Hospital, Kumamoto, Japan
| | - Tomio Jinnouchi
- Diabetes Care Center, Jinnouchi Hospital, Kumamoto, Japan
- Division of Cardiovascular Medicine, Diabetes Care Center, Jinnouchi Hospital, Kumamoto, Japan
| |
Collapse
|
33
|
Tura A, Göbl C, Vardarli I, Pacini G, Nauck M. Insulin clearance and incretin hormones following oral and "isoglycemic" intravenous glucose in type 2 diabetes patients under different antidiabetic treatments. Sci Rep 2022; 12:2510. [PMID: 35169165 PMCID: PMC8847358 DOI: 10.1038/s41598-022-06402-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 12/09/2021] [Indexed: 12/14/2022] Open
Abstract
It has not been elucidated whether incretins affect insulin clearance in type 2 diabetes (T2D). We aimed exploring possible associations between insulin clearance and endogenously secreted or exogenously administered incretins in T2D patients. Twenty T2D patients were studied (16 males/4 females, 59 ± 2 years (mean ± standard error), BMI = 31 ± 1 kg/m2, HbA1c = 7.0 ± 0.1%). Patients were treated with metformin, sitagliptin, metformin/sitagliptin combination, and placebo (randomized order). On each treatment period, oral and isoglycemic intravenous glucose infusion tests were performed (OGTT, IIGI, respectively). We also studied twelve T2D patients (9 males/3 females, 61 ± 3 years, BMI = 30 ± 1 kg/m2, HbA1c = 7.3 ± 0.4%) that underwent infusion of GLP-1(7-36)-amide, GIP, GLP-1/GIP combination, and placebo. Plasma glucose, insulin, C-peptide, and incretins were measured. Insulin clearance was assessed as insulin secretion to insulin concentration ratio. In the first study, we found OGTT/IIGI insulin clearance ratio weakly inversely related to OGTT/IIGI total GIP and intact GLP-1 (R2 = 0.13, p < 0.02). However, insulin clearance showed some differences between sitagliptin and metformin treatment (p < 0.02). In the second study we found no difference in insulin clearance following GLP-1 and/or GIP infusion (p > 0.5). Thus, our data suggest that in T2D there are no relevant incretin effects on insulin clearance. Conversely, different antidiabetic treatments may determine insulin clearance variations.
Collapse
Affiliation(s)
- Andrea Tura
- CNR Institute of Neuroscience, Corso Stati Uniti 4, 35127, Padova, Italy.
| | - Christian Göbl
- Department of Obstetrics and Gynaecology, Medical University of Vienna, Vienna, Austria
| | - Irfan Vardarli
- Diabetes Division, Katholisches Klinikum Bochum, St. Josef Hospital (Ruhr University Bochum), Bochum, Germany
| | | | - Michael Nauck
- Diabetes Division, Katholisches Klinikum Bochum, St. Josef Hospital (Ruhr University Bochum), Bochum, Germany
| |
Collapse
|
34
|
Bergman RN, Kabir M, Ader M. The Physiology of Insulin Clearance. Int J Mol Sci 2022; 23:1826. [PMID: 35163746 PMCID: PMC8836929 DOI: 10.3390/ijms23031826] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/02/2022] [Accepted: 02/02/2022] [Indexed: 02/05/2023] Open
Abstract
In the 1950's, Dr. I. Arthur Mirsky first recognized the possible importance of insulin degradation changes to the pathogenesis of type 2 diabetes. While this mechanism was ignored for decades, insulin degradation is now being recognized as a possible factor in diabetes risk. After Mirsky, the relative importance of defects in insulin release and insulin resistance were recognized as risk factors. The hyperbolic relationship between secretion and sensitivity was introduced, as was the relationship between them, as expressed as the disposition index (DI). The DI was shown to be affected by environmental and genetic factors, and it was shown to be differentiated among ethnic groups. However, the importance of differences in insulin degradation (clearance) on the disposition index relationship remains to be clarified. Direct measure of insulin clearance revealed it to be highly variable among even normal individuals, and to be affected by fat feeding and other physiologic factors. Insulin clearance is relatively lower in ethnic groups at high risk for diabetes such as African Americans and Hispanic Americans, compared to European Americans. These differences exist even for young children. Two possible mechanisms have been proposed for the importance of insulin clearance for diabetes risk: in one concept, insulin resistance per se leads to reduced clearance and diabetes risk. In a second and new concept, reduced degradation is a primary factor leading to diabetes risk, such that lower clearance (resulting from genetics or environment) leads to systemic hyperinsulinemia, insulin resistance, and beta-cell stress. Recent data by Chang and colleagues appear to support this latter hypothesis in Native Americans. The importance of insulin clearance as a risk factor for metabolic disease is becoming recognized and may be treatable.
Collapse
Affiliation(s)
- Richard N. Bergman
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (M.K.); (M.A.)
| | | | | |
Collapse
|
35
|
Okura T, Fujioka Y, Nakamura R, Kitao S, Ito Y, Anno M, Matsumoto K, Shoji K, Matsuzawa K, Izawa S, Okura H, Ueta E, Kato M, Imamura T, Taniguchi SI, Yamamoto K. The sodium-glucose cotransporter 2 inhibitor ipragliflozin improves liver function and insulin resistance in Japanese patients with type 2 diabetes. Sci Rep 2022; 12:1896. [PMID: 35115614 PMCID: PMC8814145 DOI: 10.1038/s41598-022-05704-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 01/18/2022] [Indexed: 11/26/2022] Open
Abstract
Sodium-glucose cotransporter 2 inhibitor (SGLT2i) treatment is a therapeutic approach for type 2 diabetes mellitus (T2DM). Some reports have shown that SGLT2i treatment improves insulin resistance; however, few studies have evaluated insulin resistance by the glucose clamp method. Hepatic insulin clearance (HIC) is a new pathophysiological mechanism of T2DM. The effect of SGLT2i treatment on hepatic insulin clearance and insulin resistance is not well known. We investigated the effect of SGLT2i treatment on insulin resistance, insulin secretion, incretin levels, body composition, and hepatic insulin clearance. We conducted a meal tolerance test (MTT) and a hyperinsulinemic-euglycemic clamp test in 9 T2DM patients. Ipragliflozin (50 mg/day) was administered, and the MTT and clamp test were performed after 4 months. We calculated HIC as the postprandial C-peptide AUC-to-insulin AUC ratio. We also measured GLP-1, GIP, and glucagon levels during the MTT. Body weight and HbA1c were decreased, although not significantly, after 4 months of treatment. Postprandial glucose, fasting insulin and postprandial insulin were significantly decreased. Insulin resistance with the glucose clamp was not changed, but the HOMA-IR and insulin sensitivity indices were significantly improved. Incretin and glucagon levels were not changed. Hepatic insulin clearance was significantly increased, but whole-body insulin clearance was not changed. The FIB-4 index and fatty liver index were significantly reduced. The HOMA-beta and insulinogenic indices were not changed, but the C-peptide index was significantly increased. Although the number of patients was small, these results suggested that SGLT2i treatment improved liver function, decreased hepatic insulin resistance, and increased hepatic insulin clearance, despite the small weight reduction.
Collapse
Affiliation(s)
- Tsuyoshi Okura
- Division of Cardiovascular Medicine, Endocrinology and Metabolism, Tottori University Faculty of Medicine, 36-1 Nishi-Cho, Yonago, Tottori, 683-8504, Japan.
| | - Yohei Fujioka
- Division of Cardiovascular Medicine, Endocrinology and Metabolism, Tottori University Faculty of Medicine, 36-1 Nishi-Cho, Yonago, Tottori, 683-8504, Japan
| | - Risa Nakamura
- Division of Cardiovascular Medicine, Endocrinology and Metabolism, Tottori University Faculty of Medicine, 36-1 Nishi-Cho, Yonago, Tottori, 683-8504, Japan
| | - Sonoko Kitao
- Division of Cardiovascular Medicine, Endocrinology and Metabolism, Tottori University Faculty of Medicine, 36-1 Nishi-Cho, Yonago, Tottori, 683-8504, Japan
| | - Yuichi Ito
- Division of Cardiovascular Medicine, Endocrinology and Metabolism, Tottori University Faculty of Medicine, 36-1 Nishi-Cho, Yonago, Tottori, 683-8504, Japan
| | - Mari Anno
- Division of Cardiovascular Medicine, Endocrinology and Metabolism, Tottori University Faculty of Medicine, 36-1 Nishi-Cho, Yonago, Tottori, 683-8504, Japan
| | - Kazuhisa Matsumoto
- Division of Cardiovascular Medicine, Endocrinology and Metabolism, Tottori University Faculty of Medicine, 36-1 Nishi-Cho, Yonago, Tottori, 683-8504, Japan
| | - Kyoko Shoji
- Division of Cardiovascular Medicine, Endocrinology and Metabolism, Tottori University Faculty of Medicine, 36-1 Nishi-Cho, Yonago, Tottori, 683-8504, Japan
| | - Kazuhiko Matsuzawa
- Division of Cardiovascular Medicine, Endocrinology and Metabolism, Tottori University Faculty of Medicine, 36-1 Nishi-Cho, Yonago, Tottori, 683-8504, Japan
| | - Shoichiro Izawa
- Division of Cardiovascular Medicine, Endocrinology and Metabolism, Tottori University Faculty of Medicine, 36-1 Nishi-Cho, Yonago, Tottori, 683-8504, Japan
| | - Hiroko Okura
- Division of Cardiovascular Medicine, Endocrinology and Metabolism, Tottori University Faculty of Medicine, 36-1 Nishi-Cho, Yonago, Tottori, 683-8504, Japan
| | - Etsuko Ueta
- School of Health Science, Tottori University Faculty of Medicine, Yonago, Japan
| | - Masahiko Kato
- School of Health Science, Tottori University Faculty of Medicine, Yonago, Japan
| | - Takeshi Imamura
- Division of Molecular Pharmacology, Tottori University Faculty of Medicine, Yonago, Japan
| | - Shin-Ichi Taniguchi
- Department of Regional Medicine, Tottori University Faculty of Medicine, Yonago, Japan
| | - Kazuhiro Yamamoto
- Division of Cardiovascular Medicine, Endocrinology and Metabolism, Tottori University Faculty of Medicine, 36-1 Nishi-Cho, Yonago, Tottori, 683-8504, Japan
| |
Collapse
|
36
|
Fu Z, Wu Q, Guo W, Gu J, Zheng X, Gong Y, Lu C, Ye J, Ye X, Jiang W, Hu M, Yu B, Fu Q, Liu X, Bai J, Li JZ, Yang T, Zhou H. Impaired Insulin Clearance as the Initial Regulator of Obesity-Associated Hyperinsulinemia: Novel Insight Into the Underlying Mechanism Based on Serum Bile Acid Profiles. Diabetes Care 2022; 45:425-435. [PMID: 34880066 DOI: 10.2337/dc21-1023] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 11/12/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To investigate the roles of insulin clearance and insulin secretion in the development of hyperinsulinemia in obese subjects and to reveal the association between insulin clearance and bile acids (BAs). RESEARCH DESIGN AND METHODS In cohort 1, insulin secretion, sensitivity, and endogenous insulin clearance were evaluated with an oral glucose tolerance test in 460 recruited participants. In cohort 2, 81 participants underwent an intravenous glucose tolerance test and a hyperinsulinemic-euglycemic clamp to assess insulin secretion, endogenous and exogenous insulin clearance, and insulin sensitivity. Based on insulin resistance levels ranging from mild to severe, obese participants without diabetes were further divided into 10 quantiles in cohort 1 and into tertiles in cohort 2. Forty serum BAs were measured in cohort 2 to examine the association between BAs and insulin clearance. RESULTS All obese participants had impaired insulin clearance, and it worsened with additional insulin resistance in obese subjects without diabetes. However, insulin secretion was unchanged from quantile 1 to 3 in cohort 1, and no difference was found in cohort 2. After adjustments for all confounding factors, serum-conjugated BAs, especially glycodeoxycholic acid (GDCA; β = -0.335, P = 0.004) and taurodeoxycholic acid (TDCA; β = -0.333, P = 0.003), were negatively correlated with insulin clearance. The ratio of unconjugated to conjugated BAs (β = 0.335, P = 0.002) was positively correlated with insulin clearance. CONCLUSIONS Hyperinsulinemia in obese subjects might be primarily induced by decreased insulin clearance rather than increased insulin secretion. Changes in circulating conjugated BAs, especially GDCA and TDCA, might play an important role in regulating insulin clearance.
Collapse
Affiliation(s)
- Zhenzhen Fu
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qinyi Wu
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wen Guo
- Department of Health Promotion Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jingyu Gu
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xuqin Zheng
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yingyun Gong
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chenyan Lu
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jingya Ye
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xuan Ye
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wanzi Jiang
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Moran Hu
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Baowen Yu
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qi Fu
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiang Liu
- Beijing Academy of Artificial Intelligence, Beijing, China.,College of Future Technology, Peking University, Beijing, China
| | - Jianling Bai
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - John Zhong Li
- The Key Laboratory of Rare Metabolic Disease, Department of Biochemistry and Molecular Biology, The Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Tao Yang
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hongwen Zhou
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
37
|
Chan CL, Pyle L, Vigers T, Zeitler PS, Nadeau KJ. The Relationship Between Continuous Glucose Monitoring and OGTT in Youth and Young Adults With Cystic Fibrosis. J Clin Endocrinol Metab 2022; 107:e548-e560. [PMID: 34537845 PMCID: PMC8764335 DOI: 10.1210/clinem/dgab692] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Indexed: 01/13/2023]
Abstract
CONTEXT Early glucose abnormalities in people with cystic fibrosis (PwCF) are commonly detected by continuous glucose monitoring (CGM). Relationships between these CGM abnormalities and oral glucose tolerance testing (OGTT) in PwCF have not been fully characterized. OBJECTIVE This work aimed to determine the relationship between CGM and common OGTT-derived estimates of β-cell function, including C-peptide index and oral disposition index (oDI) and to explore whether CGM can be used to screen for OGTT-defined prediabetes and cystic fibrosis-related diabetes (CFRD). METHODS PwCF not on insulin and healthy controls aged 6 to 25 years were enrolled in a prospective study collecting OGTT and CGM. A subset underwent frequently sampled OGTTs (fsOGTT) with 7-point glucose, insulin, and C-peptide measurements. Pearson correlation coefficient was used to test the association between select CGM and fsOGTT measures. Receiver operating curve (ROC) analysis was applied to CGM variables to determine the cutoff optimizing sensitivity and specificity for detecting prediabetes and CFRD. RESULTS A total of 120 participants (controls = 35, CF = 85), including 69 with fsOGTTs, were included. CGM coefficient of variation correlated inversely with C-peptide index (Cpeptide30-Cpeptide0/Glucose30-Glucose0) (r = -0.45, P < .001) and oDIcpeptide (C-peptide index)(1/cpep0) (r = -0.48, P < .0001). In PwCF, CGM variables had ROC - areas under the curve ranging from 0.43 to 0.57 for prediabetes and 0.47 to 0.6 for CFRD. CONCLUSION Greater glycemic variability on CGM correlated with reduced β-cell function. However, CGM performed poorly at discriminating individuals with and without OGTT-defined CFRD and prediabetes. Prospective studies are now needed to determine how well the different tests predict clinically relevant nonglycemic outcomes in PwCF.
Collapse
Affiliation(s)
- Christine L Chan
- Department of Pediatrics, Pediatric Endocrinology, Children’s Hospital Colorado and University of Colorado Anschutz Medical Center, Aurora, Colorado 80045, USA
| | - Laura Pyle
- Department of Biostatistics, Colorado School of Public Health, Aurora, Colorado 80045, USA
| | - Tim Vigers
- Department of Pediatrics, Pediatric Endocrinology, Children’s Hospital Colorado and University of Colorado Anschutz Medical Center, Aurora, Colorado 80045, USA
- Department of Biostatistics, Colorado School of Public Health, Aurora, Colorado 80045, USA
| | - Philip S Zeitler
- Department of Pediatrics, Pediatric Endocrinology, Children’s Hospital Colorado and University of Colorado Anschutz Medical Center, Aurora, Colorado 80045, USA
| | - Kristen J Nadeau
- Department of Pediatrics, Pediatric Endocrinology, Children’s Hospital Colorado and University of Colorado Anschutz Medical Center, Aurora, Colorado 80045, USA
| |
Collapse
|
38
|
Lee SJ, Chandrasekran P, Mazucanti CH, O’Connell JF, Egan JM, Kim Y. Dietary curcumin restores insulin homeostasis in diet-induced obese aged mice. Aging (Albany NY) 2022; 14:225-239. [PMID: 35017319 PMCID: PMC8791219 DOI: 10.18632/aging.203821] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 12/29/2021] [Indexed: 06/14/2023]
Abstract
Although aging is a physiological process to which all organisms are subject, the presence of obesity and type 2 diabetes accelerates biological aging. Recent studies have demonstrated the causal relationships between dietary interventions suppressing obesity and type 2 diabetes and delaying the onset of age-related endocrine changes. Curcumin, a natural antioxidant, has putative therapeutic properties such as improving insulin sensitivity in obese mice. However, how curcumin contributes to maintaining insulin homeostasis in aged organisms largely remains unclear. Thus, the objective of this study is to examine the pleiotropic effect of dietary curcumin on insulin homeostasis in a diet-induced obese (DIO) aged mouse model. Aged (18-20 months old) male mice given a high-fat high-sugar diet supplemented with 0.4% (w/w) curcumin (equivalent to 2 g/day for a 60 kg adult) displayed a different metabolic phenotype compared to mice given a high-fat high-sugar diet alone. Furthermore, curcumin supplementation altered hepatic gene expression profiling, especially insulin signaling and senescence pathways. We then mechanistically investigated how curcumin functions to fine-tune insulin sensitivity. We found that curcumin supplementation increased hepatic insulin-degrading enzyme (IDE) expression levels and preserved islet integrity, both outcomes that are beneficial to preserving good health with age. Our findings suggest that the multifaceted therapeutic potential of curcumin can be used as a protective agent for age-induced metabolic diseases.
Collapse
Affiliation(s)
- Su-Jeong Lee
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | - Prabha Chandrasekran
- Laboratory of Clinical Investigation, National Institute on Aging (NIA), Baltimore, MD 21224, USA
| | - Caio Henrique Mazucanti
- Laboratory of Clinical Investigation, National Institute on Aging (NIA), Baltimore, MD 21224, USA
| | - Jennifer F. O’Connell
- Laboratory of Clinical Investigation, National Institute on Aging (NIA), Baltimore, MD 21224, USA
| | - Josephine M. Egan
- Laboratory of Clinical Investigation, National Institute on Aging (NIA), Baltimore, MD 21224, USA
| | - Yoo Kim
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| |
Collapse
|
39
|
Koh HCE, Cao C, Mittendorfer B. Insulin Clearance in Obesity and Type 2 Diabetes. Int J Mol Sci 2022; 23:596. [PMID: 35054781 PMCID: PMC8776220 DOI: 10.3390/ijms23020596] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/02/2022] [Accepted: 01/03/2022] [Indexed: 02/06/2023] Open
Abstract
Plasma insulin clearance is an important determinant of plasma insulin concentration. In this review, we provide an overview of the factors that regulate insulin removal from plasma and discuss the interrelationships among plasma insulin clearance, excess adiposity, insulin sensitivity, and type 2 diabetes (T2D). We conclude with the perspective that the commonly observed lower insulin clearance rate in people with obesity, compared with lean people, is not a compensatory response to insulin resistance but occurs because insulin sensitivity and insulin clearance are mechanistically, directly linked. Furthermore, insulin clearance decreases postprandially because of the marked increase in insulin delivery to tissues that clear insulin. The commonly observed high postprandial insulin clearance in people with obesity and T2D likely results from the relatively low insulin secretion rate, not an impaired adaptation of tissues that clear insulin.
Collapse
Affiliation(s)
| | | | - Bettina Mittendorfer
- Center for Human Nutrition, Washington University School of Medicine, 660 S Euclid Ave, Campus Box 8031-14-0002, St. Louis, MO 63110, USA; (H.-C.E.K.); (C.C.)
| |
Collapse
|
40
|
Mittendorfer B, Patterson BW, Smith GI, Yoshino M, Klein S. Beta-cell function and plasma insulin clearance in people with obesity and different glycemic status. J Clin Invest 2021; 132:154068. [PMID: 34905513 PMCID: PMC8803344 DOI: 10.1172/jci154068] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 12/08/2021] [Indexed: 12/02/2022] Open
Abstract
Background It is unclear how excess adiposity and insulin resistance affect β cell function, insulin secretion, and insulin clearance in people with obesity. Methods We used a hyperinsulinemic-euglycemic clamp procedure and a modified oral glucose tolerance test to evaluate the interrelationships among obesity, insulin sensitivity, insulin kinetics, and glycemic status in 5 groups of individuals: normoglycemic lean and obese individuals with (a) normal fasting glucose and normal glucose tolerance (Ob-NFG-NGT), (b) NFG and impaired glucose tolerance (Ob-NFG-IGT), (c) impaired fasting glucose and IGT (Ob-IFG-IGT), or (d) type 2 diabetes (Ob-T2D). Results Glucose-stimulated insulin secretion (GSIS), an assessment of β cell function, was greater in the Ob-NFG-NGT and Ob-NFG-IGT groups than in the lean group, even when insulin sensitivity was matched in the obese and lean groups. Insulin sensitivity, not GSIS, was decreased in the Ob-NFG-IGT group compared with the Ob-NFG-NGT group, whereas GSIS, not insulin sensitivity, was decreased in the Ob-IFG-IGT and Ob-T2D groups compared with the Ob-NFG-NGT and Ob-NFG-IGT groups. Insulin clearance was directly related to insulin sensitivity and inversely related to the postprandial increase in insulin secretion and plasma insulin concentration. Conclusion Increased adiposity per se, not insulin resistance, enhanced insulin secretion in people with obesity. The obesity-induced increase in insulin secretion, in conjunction with a decrease in insulin clearance, sufficiently raised the plasma insulin concentrations needed to maintain normoglycemia in individuals with moderate, but not severe, insulin resistance. A deterioration in β cell function, not a decrease in insulin sensitivity, was a determinant of IFG and ultimately leads to T2D. CLINICAL TRIALS REGISTRATION ClinicalTrials.gov NCT02706262, NCT04131166, and NCT01977560. FUNDING NIH (P30 DK056341, P30 DK020579, and UL1 TR000448); American Diabetes Association (1-18-ICTS-119); Longer Life Foundation; Pershing Square Foundation; and Washington University-Centene ARCH Personalized Medicine Initiative (P19-00559).
Collapse
Affiliation(s)
- Bettina Mittendorfer
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, United States of America
| | - Bruce W Patterson
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, United States of America
| | - Gordon I Smith
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, United States of America
| | - Mihoko Yoshino
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, United States of America
| | - Samuel Klein
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, United States of America
| |
Collapse
|
41
|
Piersanti A, Abdul Rahman NHB, Gobl C, Burattini L, Kautzky-Willer A, Pacini G, Tura A, Morettini M. Model-Based Assessment of Hepatic and Extrahepatic Insulin Clearance from Short Insulin-Modified IVGTT in Women with a History of Gestational Diabetes. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2021; 2021:4311-4314. [PMID: 34892175 DOI: 10.1109/embc46164.2021.9630405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Insulin clearance is an integral component of insulin metabolism. Yet, little is known about separate contribution of hepatic and extrahepatic insulin clearance in type 2 diabetes and in high-risk populations, such as women who experienced gestational diabetes mellitus (pGDM). A model-based method was recently proposed to assess both contributions from 3-hour insulin-modified intravenous glucose tolerance test (IM-IVGTT); the aim of this study was to assess the reliability of short (1 hour) IM-IVGTT in the application of such model-based method and to evaluate the role of the two contributions in determining insulin clearance in pGDM. A total of 115 pGDM women and 41 who remained healthy during pregnancy (CNT) were analyzed early postpartum and underwent a 3-hour IMIVGTT. Peripheral insulin clearance (CLP), hepatic fractional extraction (FEL) and extrahepatic distribution volume (VP) were estimated by performing a best-fit procedure on insulin IMIVGTT data considering firstly the overall 3-hour duration and then limiting data to 1 hour. Results showed no significant difference in parameter values between the 3-hour and the 1-hour IM-IVGTT. Comparison between pGDM and CNT (1-hour) showed no significant difference in CLp (0.23 [0.29] vs. 0.27 [0.43] L·min-1; p=0.64), FEL (50.2 [15.1] vs. 50.9 [11.7] %; p=0.63) and VP (2.01 [2.99] vs. 2.70 [4.00] L; p=0.92). In conclusion, short IM-IVGTT provides a reliable assessment of hepatic and extrahepatic insulin clearance through such model-based method. Its application to the study of pGDM women showed no alteration in hepatic and extrahepatic contributions with respect to women who had a healthy pregnancy.Clinical Relevance- This study proves the reliability of short (1 hour) IM-IVGTT to assess hepatic and extrahepatic insulin clearance in women who experienced gestational diabetes.
Collapse
|
42
|
Pan K, Shi X, Liu K, Wang J, Chen Y. Efficacy, Pharmacokinetics, Biodistribution and Excretion of a Novel Acylated Long-Acting Insulin Analogue INS061 in Rats. Drug Des Devel Ther 2021; 15:3487-3498. [PMID: 34408401 PMCID: PMC8364340 DOI: 10.2147/dddt.s317327] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 07/23/2021] [Indexed: 12/17/2022] Open
Abstract
Purpose Long-acting insulin analogues are known to be a major player in the management of glucose levels in type I diabetic patients. However, highly frequent hypo- and hyperglycemic incidences of current long-acting insulins are the important factor to limit stable management of glucose level for clinical benefits. To further optimize the properties for steadily controlling glucose level, a novel long-acting insulin INS061 was designed and its efficacy, pharmacokinetics, biodistribution and excretion profiles were investigated in rats. Methods The glucose-lowering effects were evaluated in a streptozocin-induced diabetic rats compared to commercial insulins via subcutaneous administration. The pharmacokinetics, biodistribution, and excretion were examined by validated analytical methods including radioactivity assay and radioactivity assay after the precipitation with TCA and the separation by HPLC. Results INS061 exhibited favorable blood glucose lowering effects up to 24 h compared to Degludec. Pharmacokinetic study revealed that the concentration-time curves of INS061 between two administration routes were remarkably different. Following intravenous administration, INS061 was quickly distributed to various organs and tissues and slowly eliminated over time with urinary excretion being the major route for elimination, and the maximum plasma concentrations (Cmax) and systemic exposures (AUC) increased in a linear manner. Conclusion The present structural modifications of human insulin possessed a long-acting profile and glucose-lowering function along with favorable in vivo properties in rats, which establish a foundation for further preclinical and clinical evaluation.
Collapse
Affiliation(s)
- Kai Pan
- State Key Laboratory of Natural Medicines and Laboratory of Chemical Biology, China Pharmaceutical University, Nanjing, Jiangsu Province, 211198, People's Republic of China.,Jiangsu Hengrui Medicine Co., Ltd., Lianyungang, 222047, People's Republic of China
| | - Xiaolei Shi
- Jiangsu Hengrui Medicine Co., Ltd., Lianyungang, 222047, People's Republic of China
| | - Kai Liu
- Fujian Suncadia Medicine Co., Ltd, Xiamen, 361026, People's Republic of China
| | - Ju Wang
- Jiangsu Hengrui Medicine Co., Ltd., Lianyungang, 222047, People's Republic of China
| | - Yijun Chen
- State Key Laboratory of Natural Medicines and Laboratory of Chemical Biology, China Pharmaceutical University, Nanjing, Jiangsu Province, 211198, People's Republic of China
| |
Collapse
|
43
|
Galderisi A, Moran A, Evans-Molina C, Martino M, Santoro N, Caprio S, Cobelli C. Early Impairment of Insulin Sensitivity, β-Cell Responsiveness, and Insulin Clearance in Youth with Stage 1 Type 1 Diabetes. J Clin Endocrinol Metab 2021; 106:2660-2669. [PMID: 34000022 PMCID: PMC8372628 DOI: 10.1210/clinem/dgab344] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Indexed: 01/10/2023]
Abstract
CONTEXT Clinical onset of type 1 diabetes (Stage 3 T1D) is preceded by a presymptomatic phase characterized by multiple islet autoantibodies with normal glucose tolerance (Stage 1 T1D). OBJECTIVE The aim was to explore the metabolic phenotypes of β-cell function and insulin sensitivity and clearance in normoglycemic youth with Stage 1 T1D and compare them with healthy nonrelated peers during a 3-hour oral glucose tolerance test (OGTT). METHODS Twenty-eight lean youth, 14 with ≥2 islet autoantibodies (cases) and 14 healthy controls underwent a 3-hour 9-point OGTT with measurement of glucose, C-peptide, and insulin. The oral minimal model was used to quantitate β-cell responsiveness (φtotal) and insulin sensitivity (SI), allowing assessment of β-cell function by the disposition index (DI=φtotal×SI). Fasting insulin clearance (CL0) was calculated as the ratio between the fasting insulin secretion rate (ISR) and plasma insulin levels (ISR0/I0), while postload clearance (CL180) was estimated by the ratio of AUC of ISR over the plasma insulin AUC for the 3-hour OGTT (ISRAUC/IAUC). Participants with impaired fasting glucose, impaired glucose tolerance, or any OGTT glucose concentration ≥200 mg/dL were excluded. RESULTS Cases (10.5 years [8, 15]) exhibited reduced DI (P < .001) due to a simultaneous reduction in both φtotal (P < 0.001) and SI (P = .008) compared with controls (11.5 years [10.4, 14.9]). CL0 and CL180 were lower in cases than in controls (P = .005 and P = .019). CONCLUSION Presymptomatic Stage 1 T1D in youth is associated with reduced insulin sensitivity and lower β-cell responsiveness, and the presence of blunted insulin clearance.
Collapse
Affiliation(s)
- Alfonso Galderisi
- Department of Woman and Child’s Health, University of Padova, Padova, Italy
- Department of Pediatrics, Yale University, New Haven, CT, USA
- Correspondence: Alfonso Galderisi, MD, PhD, Department of Woman and Child’s Health, University of Padova, Via N. Giustiniani, 3, 35128 Padova, Italy.
| | - Antoinette Moran
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Carmella Evans-Molina
- Center for Diabetes and Metabolic Diseases, Indiana University, Bloomington, IN, USA
| | - Mariangela Martino
- Department of Woman and Child’s Health, University of Padova, Padova, Italy
| | - Nicola Santoro
- Department of Pediatrics, Yale University, New Haven, CT, USA
- Department of Medicine and Health Sciences “V. Tiberio,” University of Molise, Campobasso, Italy
| | - Sonia Caprio
- Department of Pediatrics, Yale University, New Haven, CT, USA
| | - Claudio Cobelli
- Department of Woman and Child’s Health, University of Padova, Padova, Italy
| |
Collapse
|
44
|
Wood AC, Jensen ET, Bertoni AG, Ramesh G, Rich SS, Rotter JI, Chen YDI, Goodarzi MO. Defining the Relative Role of Insulin Clearance in Early Dysglycemia in Relation to Insulin Sensitivity and Insulin Secretion: The Microbiome and Insulin Longitudinal Evaluation Study (MILES). Metabolites 2021; 11:420. [PMID: 34206745 PMCID: PMC8304591 DOI: 10.3390/metabo11070420] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 06/07/2021] [Accepted: 06/16/2021] [Indexed: 11/30/2022] Open
Abstract
Insulin resistance and insufficient insulin secretion are well-recognized contributors to type 2 diabetes. A potential role of reduced insulin clearance has been suggested, but few studies have investigated the contribution of insulin clearance while simultaneously examining decreased insulin sensitivity and secretion. The goal of this study was to conduct such an investigation in a cohort of 353 non-Hispanic White and African American individuals recruited in the Microbiome and Insulin Longitudinal Evaluation Study (MILES). Participants underwent oral glucose tolerance tests from which insulin sensitivity, insulin secretion, insulin clearance, and disposition index were calculated. Regression models examined the individual and joint contributions of these traits to early dysglycemia (prediabetes or newly diagnosed diabetes). In separate models, reduced insulin sensitivity, reduced disposition index, and reduced insulin clearance were associated with dysglycemia. In a joint model, only insulin resistance and reduced insulin secretion were associated with dysglycemia. Models with insulin sensitivity, disposition index, or three insulin traits had the highest discriminative value for dysglycemia (area under the receiver operating characteristics curve of 0.82 to 0.89). These results suggest that in the race groups studied, insulin resistance and compromised insulin secretion are the main independent underlying defects leading to early dysglycemia.
Collapse
Affiliation(s)
- Alexis C. Wood
- USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Elizabeth T. Jensen
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA; (E.T.J.); (A.G.B.)
| | - Alain G. Bertoni
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA; (E.T.J.); (A.G.B.)
| | - Gautam Ramesh
- School of Medicine, University of California, La Jolla, San Diego, CA 92093, USA;
| | - Stephen S. Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA 22908, USA;
| | - Jerome I. Rotter
- Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (J.I.R.); (Y.-D.I.C.)
| | - Yii-Der I. Chen
- Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (J.I.R.); (Y.-D.I.C.)
| | - Mark O. Goodarzi
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
45
|
Laurenti MC, Matveyenko A, Vella A. Measurement of Pulsatile Insulin Secretion: Rationale and Methodology. Metabolites 2021; 11:409. [PMID: 34206296 PMCID: PMC8305896 DOI: 10.3390/metabo11070409] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/17/2021] [Accepted: 06/18/2021] [Indexed: 12/29/2022] Open
Abstract
Pancreatic β-cells are responsible for the synthesis and exocytosis of insulin in response to an increase in circulating glucose. Insulin secretion occurs in a pulsatile manner, with oscillatory pulses superimposed on a basal secretion rate. Insulin pulses are a marker of β-cell health, and secretory parameters, such as pulse amplitude, time interval and frequency distribution, are impaired in obesity, aging and type 2 diabetes. In this review, we detail the mechanisms of insulin production and β-cell synchronization that regulate pulsatile insulin secretion, and we discuss the challenges to consider when measuring fast oscillatory secretion in vivo. These include the anatomical difficulties of measuring portal vein insulin noninvasively in humans before the hormone is extracted by the liver and quickly removed from the circulation. Peripheral concentrations of insulin or C-peptide, a peptide cosecreted with insulin, can be used to estimate their secretion profile, but mathematical deconvolution is required. Parametric and nonparametric approaches to the deconvolution problem are evaluated, alongside the assumptions and trade-offs required for their application in the quantification of unknown insulin secretory rates from known peripheral concentrations. Finally, we discuss the therapeutical implication of targeting impaired pulsatile secretion and its diagnostic value as an early indicator of β-cell stress.
Collapse
Affiliation(s)
- Marcello C. Laurenti
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic, Rochester, MN 55905, USA; (M.C.L.); (A.M.)
- Biomedical Engineering and Physiology Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA
| | - Aleksey Matveyenko
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic, Rochester, MN 55905, USA; (M.C.L.); (A.M.)
| | - Adrian Vella
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic, Rochester, MN 55905, USA; (M.C.L.); (A.M.)
| |
Collapse
|
46
|
Bergman RN, Piccinini F. Response to Comment on Piccinini and Bergman The Measurement of Insulin Clearance. Diabetes Care 2020;43:2296-2302. Diabetes Care 2021; 44:e100-e101. [PMID: 33972319 PMCID: PMC8132327 DOI: 10.2337/dci20-0080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
47
|
Gastaldelli A, DeFronzo RA, Salehi M. Comment on Piccinini and Bergman. The Measurement of Insulin Clearance. Diabetes Care 2020;43:2296-2302. Diabetes Care 2021; 44:e98-e99. [PMID: 33972318 PMCID: PMC8132326 DOI: 10.2337/dc20-2911] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Amalia Gastaldelli
- University of Texas Health Science Center, San Antonio, TX
- Institute of Clinical Physiology, CNR, Pisa, Italy
| | | | - Marzieh Salehi
- University of Texas Health Science Center, San Antonio, TX
- Audie L. Murphy Memorial Veterans' Hospital, South Texas Veterans Health Care System, San Antonio, TX
| |
Collapse
|
48
|
Chen M, Zhu B, Chen D, Hu X, Xu X, Shen WJ, Hu C, Li J, Qu S. COVID-19 May Increase the Risk of Insulin Resistance in Adult Patients Without Diabetes: A 6-Month Prospective Study. Endocr Pract 2021; 27:834-841. [PMID: 33887468 PMCID: PMC8054613 DOI: 10.1016/j.eprac.2021.04.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 04/02/2021] [Accepted: 04/08/2021] [Indexed: 02/07/2023]
Abstract
OBJECTIVE During the coronavirus disease 2019 (COVID-19) pandemic, exploring insulin resistance and beta-cell activity is important for understanding COVID-19‒associated new-onset diabetes. We assessed insulin sensitivity and fasting insulin secretion in patients with COVID-19 without diabetes on admission and at 3 and 6 months after discharge. METHODS This 6-month prospective study assessed data from the records of 64 patients without diabetes diagnosed with COVID-19 at Wenzhou Central Hospital, China. Each patient was followed up at 3 and 6 months after discharge. Repeated measures analysis of variance was used to investigate differences in multiple measurements of the same variable at different times. Linear regression analysis was performed to analyze the contributor for changes in the triglyceride-glucose (TyG) index. RESULTS Fasting C-peptide levels in patients at baseline were lower than the normal range. Compared with the baseline results, patients had significantly elevated fasting C-peptide levels (0.35 ± 0.24 vs 2.36 ± 0.98 vs 2.52 ± 1.11 μg/L; P < .001), homeostasis model assessment for beta-cell function (0.42, interquartile range [IQR] 0.36-0.62 vs 2.54, IQR 1.95-3.42 vs 2.90, IQR 2.02-4.23; P < .001), and TyG indices (8.57 ± 0.47 vs 8.73 ± 0.60 vs 8.82 ± 0.62; P = .006) and decreased fasting glucose levels (5.84 ± 1.21 vs 4.95 ± 0.76 vs 5.40 ± 0.68 mmol/L; P = .003) at the 3- and 6-month follow-up. Male gender, age, interferon-alfa treatment during hospitalization, and changes in total cholesterol and high-density lipoprotein levels were significantly associated with changes in the TyG index. CONCLUSION Our study provided the first evidence that COVID-19 may increase the risk of insulin resistance in patients without diabetes.
Collapse
Affiliation(s)
- Mochuan Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Bing Zhu
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Dong Chen
- Department of Infectious Disease, The Ding Li Clinical College of Wenzhou Medical University and Sixth People's Hospital of Wenzhou, Wenzhou, Zhejiang, China
| | - Xingzhong Hu
- Department of Infectious Disease, The Ding Li Clinical College of Wenzhou Medical University and Sixth People's Hospital of Wenzhou, Wenzhou, Zhejiang, China
| | - Xueqin Xu
- Department of Infectious Disease, The Ding Li Clinical College of Wenzhou Medical University and Sixth People's Hospital of Wenzhou, Wenzhou, Zhejiang, China
| | - Wen-Jun Shen
- Division of Endocrinology, Gerontology and Metabolism, Stanford University School of Medicine, Stanford, California; Geriatric Research, Education, and Clinical Center, VA Palo Alto Health Care System, Palo Alto, California
| | - Chenchan Hu
- Department of Infectious Disease, The Ding Li Clinical College of Wenzhou Medical University and Sixth People's Hospital of Wenzhou, Wenzhou, Zhejiang, China.
| | - Jue Li
- Shanghai Pudong New Area Mental Health Center, Tongji University School of Medicine, Shanghai, China.
| | - Shen Qu
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
49
|
Taschetto APD, Zimath PL, Silvério R, Dos Santos C, Boschero AC, Dos Santos GJ, Rafacho A. Reduced insulin sensitivity and increased β/α cell mass is associated with reduced hepatic insulin-degrading enzyme activity in pregnant rats. Life Sci 2021; 277:119509. [PMID: 33865877 DOI: 10.1016/j.lfs.2021.119509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/05/2021] [Accepted: 04/07/2021] [Indexed: 11/19/2022]
Abstract
AIMS Pregnancy is associated with the development of a transitory insulin resistance that parallels with the upregulation of pancreatic β-cell function and mass. These metabolic adaptations guarantee the higher insulin demand, but there is no evidence of whether insulin clearance contributes to this process. Thus, we investigated some of the hepatic parameters related to insulin clearance during rat pregnancy. We also investigated some molecular parameters in the hypothalamus. MAIN METHODS We evaluated the body mass and food intake, insulin sensitivity, β- and α-cell masses, insulin clearance based on an exogenous insulin load, hepatic insulin-degrading enzyme (IDE) activity, and hepatic and hypothalamic protein content of IDE and carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM-1) in three periods of gestation in Wistar rats. KEY FINDINGS In the first week of pregnancy, both insulin sensitivity and clearance increased, a pattern that inverted in the third week of gestation (reduced insulin sensitivity and clearance). Diminished insulin clearance was associated with lower hepatic IDE activity and higher pancreatic β- and α-cell masses. No alteration in the hepatic IDE and CEACAM protein content was observed throughout pregnancy, but hypothalamic IDE protein content was significantly reduced in the late gestation period. SIGNIFICANCE In conclusion, elevated insulin demand in the late period of gestation occurs not only as a result of increased β-cell mass and function but also by a potential reduction in hepatic insulin clearance. Knowing this physiological process may be valuable when considering gestational diabetes mellitus results from a failure in insulin supply during pregnancy.
Collapse
Affiliation(s)
- Ana P D Taschetto
- Laboratory of Investigation in Chronic Diseases - LIDoC, Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina - UFSC, 88040-900 Florianópolis, Brazil; Multicenter Graduate Program in Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina - UFSC, 88040-900 Florianópolis, Brazil
| | - Priscila L Zimath
- Laboratory of Investigation in Chronic Diseases - LIDoC, Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina - UFSC, 88040-900 Florianópolis, Brazil; Graduate Program in Pharmacology, Center of Biological Sciences, Federal University of Santa Catarina - UFSC, 88040-900 Florianópolis, Brazil
| | - Renata Silvério
- Laboratory of Investigation in Chronic Diseases - LIDoC, Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina - UFSC, 88040-900 Florianópolis, Brazil; Graduate Program in Pharmacology, Center of Biological Sciences, Federal University of Santa Catarina - UFSC, 88040-900 Florianópolis, Brazil
| | - Cristiane Dos Santos
- Laboratory of Investigation in Chronic Diseases - LIDoC, Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina - UFSC, 88040-900 Florianópolis, Brazil; Graduate Program in Molecular and Functional Biology, Institute of Biology, Campinas State University - UNICAMP, 13083-862 Campinas, Brazil
| | - Antonio C Boschero
- Graduate Program in Molecular and Functional Biology, Institute of Biology, Campinas State University - UNICAMP, 13083-862 Campinas, Brazil
| | - Gustavo J Dos Santos
- Laboratory of Investigation in Chronic Diseases - LIDoC, Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina - UFSC, 88040-900 Florianópolis, Brazil; Multicenter Graduate Program in Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina - UFSC, 88040-900 Florianópolis, Brazil
| | - Alex Rafacho
- Laboratory of Investigation in Chronic Diseases - LIDoC, Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina - UFSC, 88040-900 Florianópolis, Brazil; Multicenter Graduate Program in Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina - UFSC, 88040-900 Florianópolis, Brazil; Graduate Program in Pharmacology, Center of Biological Sciences, Federal University of Santa Catarina - UFSC, 88040-900 Florianópolis, Brazil; Graduate Program in Molecular and Functional Biology, Institute of Biology, Campinas State University - UNICAMP, 13083-862 Campinas, Brazil.
| |
Collapse
|
50
|
Shah MH, Piaggi P, Looker HC, Paddock E, Krakoff J, Chang DC. Lower insulin clearance is associated with increased risk of type 2 diabetes in Native Americans. Diabetologia 2021; 64:914-922. [PMID: 33404681 DOI: 10.1007/s00125-020-05348-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/22/2020] [Indexed: 01/08/2023]
Abstract
AIMS/HYPOTHESIS Impaired insulin clearance is implicated in the pathogenesis of type 2 diabetes, but prospective evidence remains limited. Therefore, we sought to identify factors associated with the metabolic clearance rate of insulin (MCRI) and to investigate whether lower MCRI is associated with increased risk of incident type 2 diabetes. METHODS From a longitudinal cohort, 570 adult Native Americans without diabetes living in the Southwestern United States were characterised at baseline and 448 participants were monitored over a median follow-up period of 7.9 years with 146 (32%) incident cases of diabetes identified (fasting plasma glucose ≥7.0 mmol/l, 2 h plasma glucose [2-h PG] ≥11.1 mmol/l, or clinical diagnosis). At baseline, participants underwent dual-energy x-ray absorptiometry or hydrodensitometry to assess body composition, a 75 g OGTT, an IVGTT to assess acute insulin response (AIR), and a hyperinsulinaemic-euglycaemic clamp to assess MCRI and insulin action (M). RESULTS In adjusted linear models, MCRI was inversely associated with body fat percentage (r = -0.35), fasting plasma insulin (r = -0.55) and AIR (r = -0.22), and positively associated with M (r = 0.17; all p < 0.0001). In multivariable Cox proportional hazard models, lower MCRI was associated with an increased risk of diabetes after adjustment for age, sex, heritage, body fat percentage, AIR, M, fasting plasma glucose, 2-h PG, and fasting plasma insulin (HR per one-SD difference in MCRI: 0.77; 95% CI 0.61, 0.98; p = 0.03). CONCLUSIONS/INTERPRETATION Lower MCRI is associated with an unfavourable metabolic phenotype and is associated with incident type 2 diabetes independent of established risk factors. CLINICAL TRIAL REGISTRATION NUMBERS ClinicalTrials.gov NCT00339482; NCT00340132.
Collapse
Affiliation(s)
- Mujtaba H Shah
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA
| | - Paolo Piaggi
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA
| | - Helen C Looker
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA
| | - Ethan Paddock
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA
| | - Jonathan Krakoff
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA
| | - Douglas C Chang
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA.
| |
Collapse
|