1
|
Gower BA, Goss AM, Yurchishin ML, Deemer SE, Sunil B, Garvey WT. Effects of a Carbohydrate-Restricted Diet on β-Cell Response in Adults With Type 2 Diabetes. J Clin Endocrinol Metab 2025; 110:1811-1817. [PMID: 39436786 DOI: 10.1210/clinem/dgae670] [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: 06/17/2024] [Indexed: 10/25/2024]
Abstract
CONTEXT β-Cell response to glucose is compromised in individuals with type 2 diabetes (T2D), possibly due in part to excessive carbohydrate consumption. OBJECTIVE This study was conducted to determine if a eucaloric carbohydrate-restricted (CR) diet (∼9% energy from carbohydrate, 65% energy from fat), compared to a eucaloric higher carbohydrate (HC) diet (∼55% energy from carbohydrate, 20% energy from fat), would improve β-cell response to glucose in participants with T2D. METHODS Participants were 57 African American and European American adults with T2D not using insulin. Medications were discontinued 1 to 2 weeks prior to baseline testing. A hyperglycemic clamp was used to assess the acute (first-phase) and maximal (arginine-stimulated) C-peptide response to glucose at baseline and after 12 weeks of controlled diet therapy (all food provided). An oral glucose tolerance test (OGTT) was used to assess the disposition index (DI). RESULTS At 12 weeks, a statistically significant effect of diet was observed on acute C-peptide response (2-fold greater with the CR diet; P < .01). For maximal C-peptide, a significant effect of diet was observed (22% greater with the CR diet; P < .05), as was a significant diet-by-race interaction (P < .05), indicating that the diet effect was specific to European Americans (48% greater with the CR diet; P < .01). OGTT results showed a significant effect of diet on DI at 12 weeks (32% greater with the CR diet; P < .05). CONCLUSION These results suggest that a eucaloric CR diet has beneficial effects on β-cell function in patients with mild T2D.
Collapse
Affiliation(s)
- Barbara A Gower
- Department of Nutrition Sciences, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Amy M Goss
- Department of Nutrition Sciences, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Marian L Yurchishin
- Department of Nutrition Sciences, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Sarah E Deemer
- Department of Nutrition Sciences, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Bhuvana Sunil
- Division of Pediatric Endocrinology, Children's of Alabama, Birmingham, AL 35294, USA
| | - William T Garvey
- Department of Nutrition Sciences, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
2
|
Luk AOY, Wu H, Fan Y, Fan B, O CK, Chan JCN. Young-onset type 2 diabetes-Epidemiology, pathophysiology, and management. J Diabetes Investig 2025. [PMID: 40411309 DOI: 10.1111/jdi.70081] [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: 04/10/2025] [Revised: 05/22/2025] [Accepted: 05/09/2025] [Indexed: 05/26/2025] Open
Abstract
The prevalence and incidence of young-onset type 2 diabetes is increasing globally, especially in low- and middle-income countries, and predominantly affects non-White ethnic and racial populations. Young-onset type 2 is heterogeneous in terms of the genetic and environmental contributions to its underlying pathophysiology, which poses challenges for glycemic management. Young at-risk individuals remain underrepresented in clinical trials, including diabetes prevention studies, and there is still an insufficient evidence base to inform practice for this age group. Improvements in diabetes care delivery have not reached young people who will progress to have disabling complications at an age when they are most productive. This review summarizes recent studies on the epidemiology of young-onset type 2 diabetes and its complications. We discuss the genetic and environmental risk factors that act in concert to promote glycemic dysregulation and early onset of type 2 diabetes. We provide perspectives on diabetes prevention and management, and propose strategies to address the unique medical and psychosocial issues associated with young-onset type 2 diabetes. The Precision Medicine to Redefine Insulin Secretion and Monogenic Diabetes Randomized Controlled Trial (PRISM-RCT) is the first large-scale clinical trial designed to evaluate the effect of a structured care model that integrates biogenetic markers with communication and information technology on attaining strict metabolic targets and improving clinical outcomes in individuals with young-onset type 2 diabetes. The results of this study will inform the scientific community about the impact of multifactorial intervention and precision care in young patients, for whom the legacy effect is particularly significant.
Collapse
Affiliation(s)
- 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
- Hong Kong Institute of Diabetes and Obesity, 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
| | - Hongjiang Wu
- Department of Medicine and Therapeutics, 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
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Yingnan Fan
- Department of Medicine and Therapeutics, 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
- Li Ka Shing Institute of Health Sciences, 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
- Hong Kong Institute of Diabetes and Obesity, 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
| | - Chun Kwan O
- Department of Medicine and Therapeutics, 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
- Li Ka Shing Institute of Health Sciences, 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
- Hong Kong Institute of Diabetes and Obesity, 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
| |
Collapse
|
3
|
Peng XV, Klingensmith G, Hsia DS, Xie Y, Czerniak R, Tamborlane WV, Shah AS. A Randomized Phase 3 Study Evaluating the Efficacy and Safety of Alogliptin in Pediatric Participants with Type 2 Diabetes Mellitus. Diabetes Ther 2025; 16:865-883. [PMID: 40032809 PMCID: PMC12006607 DOI: 10.1007/s13300-025-01700-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/22/2025] [Indexed: 03/05/2025] Open
Abstract
INTRODUCTION There is an unmet need for pharmacological therapies for children with type 2 diabetes mellitus (T2DM). We assessed the efficacy and safety of an oral dipeptidyl peptidase-4 inhibitor, alogliptin, 25 mg once daily (QD), as a potential treatment for pediatric patients with T2DM. METHODS This phase 3, 52-week, multicenter, randomized, double-blind, placebo-controlled trial was conducted in children and adolescents (10-17 years old) with T2DM. Participants had glycosylated hemoglobin (HbA1c) ≥ 6.5% at baseline (≥ 6.5% to < 11% without treatment or on metformin alone; ≥ 7.0% to < 11% on insulin alone or in combination with metformin). Where required, participants underwent prerandomization stabilization of their background metformin and/or insulin therapy. All received diabetes education and home glucose-monitoring training (during screening, prerandomization stabilization, and specified visits through week 26). Participants were then stratified based on previous antihyperglycemic therapy for 12 weeks before screening into schedule A (antihyperglycemic treatment-naïve) or B (metformin and/or insulin). The primary efficacy endpoint was change in HbA1c levels from baseline at week 26. Safety was assessed as a secondary endpoint at week 52. RESULTS Overall, 152 participants (median age, 14 years; 68.9% female) were randomized (1:1) to receive either alogliptin (n = 75) or placebo (n = 77). The majority were white (58.3%), had a body mass index of ≥ 30 kg/m2 (60.3%), and had received previous antihyperglycemic therapy (82.1%). The difference in HbA1c levels from baseline to week 26 between the alogliptin and placebo groups (least squares mean change [95% confidence interval]) was 0.10 (- 0.63, 0.83; p = 0.78). There was no difference in efficacy endpoints between alogliptin and placebo across both subgroups. No new safety concerns were observed with alogliptin treatment. CONCLUSION Alogliptin 25 mg QD did not significantly improve glycemic control versus placebo in pediatric patients with T2DM. Alogliptin treatment was safe and well tolerated, and no new safety concerns were observed in this study. TRIAL REGISTRATION ClinicalTrials.gov: NCT02856113; EudraCT: 2015-000208-25.
Collapse
Affiliation(s)
- Xuejun Victor Peng
- Marketed Products Development, Global Portfolio Division, Takeda Pharmaceuticals Company Limited, Cambridge, MA, USA
| | | | - Daniel S Hsia
- Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Yunlong Xie
- Statistics and Quantitative Science, Takeda Pharmaceuticals Company Limited, Cambridge, MA, USA
| | - Richard Czerniak
- Quantitative Clinical Pharmacology, Takeda Pharmaceuticals Company Limited, Cambridge, MA, USA
| | - William V Tamborlane
- Department of Pediatrics and General Clinical Research Center, Yale University School of Medicine, New Haven, CT, USA
| | - Amy S Shah
- Division of Pediatric Endocrinology, Cincinnati Children's Hospital Medical Center, The University of Cincinnati, 3333 Burnet Ave, ML 7012, Cincinnati, OH, 45229, USA.
| |
Collapse
|
4
|
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
|
5
|
Shah AS, Kelsey MM, Wolf RM, Nadeau KJ. Shaping the future of youth-onset type 2 diabetes: a call to action. Trends Endocrinol Metab 2025:S1043-2760(25)00048-7. [PMID: 40187960 DOI: 10.1016/j.tem.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 04/07/2025]
Abstract
Youth-onset type 2 diabetes (YO-T2D) is an urgent public health challenge that demands immediate and innovative action. The devastating trajectory of this disease - from rapid β-cell decline to early complications and poor responses to medications - compels us to rethink our approach. Here, we argue that by investing in targeted research to unravel the unique mechanisms of this condition, ensuring equitable access to cutting-edge clinical trials, and building clinical care models tailored specifically for youth, we can rewrite the narrative for these at-risk youth.
Collapse
Affiliation(s)
- Amy S Shah
- Cincinnati Children's Hospital Medical Center and the University of Cincinnati, Department of Pediatrics, Division of Endocrinology, Cincinnati, OH, USA.
| | - Megan M Kelsey
- Section of Endocrinology, Department of Pediatrics, School of Medicine and Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Risa M Wolf
- Department of Pediatrics, Division of Endocrinology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Kristen J Nadeau
- Section of Endocrinology, Department of Pediatrics, School of Medicine and Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
6
|
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
|
7
|
Dobbs TJ, Cree MG, Bailey AJ, Baumgartner AD, Garrish J, Diniz-Behn C, Pyle L, Kelsey MM, Shah AS, Inge TH, Bjornstad P, Nadeau KJ. Glycemia, Insulin Sensitivity, and Secretion Improve 3 Months Post-sleeve Gastrectomy in Youth With Type 2 Diabetes. J Endocr Soc 2025; 9:bvaf020. [PMID: 39911520 PMCID: PMC11795195 DOI: 10.1210/jendso/bvaf020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Indexed: 02/07/2025] Open
Abstract
Context Metabolic bariatric surgery reduces weight in youth with severe obesity; however, its impacts on youth-onset type 2 diabetes (T2D) are unclear. Objective We evaluated short-term outcomes in youth with T2D 3 months after vertical sleeve gastrectomy (VSG). Design Longitudinal, observational study in the Impact of Metabolic surgery on Pancreatic, Renal, and cardiOVascular hEalth in youth with T2D study (IMPROVE-T2D). Setting Academic medical university and children's hospital. Participants Fourteen youth with T2D [mean age ± SD 16.8 ± 1.4 years; 50% female, pre-VSG hemoglobin A1c (HbA1c) 6.6 ± 0.2%; diabetes duration 17.6 ± 13.8 months; age at diabetes diagnosis 15.9 ± 1.4 years; body mass index (BMI) 46.7 ± 2 kg/m2]. Interventions Participants underwent a mixed-meal tolerance test (MMTT), body composition, and indirect calorimetry before and 3 months after VSG. Main Outcomes Glycemic control (HbA1c, diabetes medications), insulin sensitivity (Matsuda Index, Homeostasis Model of Insulin Sensitivity, oral minimal model), and secretion (C-peptide model). Results After VSG, weight and BMI decreased (25.2 ± 5.6 kg [19%], -8.7 ± 2 kg/m2 [18%], respectively, P < .001). Body fat decreased (4.5%, P = .012), with reductions of 14.1 ± 5.4 kg of fat mass (P = .005) and 4.5 kg of fat-free mass (P = .034). HbA1c decreased from 6.6 ± 0.2% to 5.7 ± 0.2% (P = .003), with 86% of participants no longer requiring diabetes medications. Glucose was lower throughout the MMTT, with insulin, C-peptide, free fatty acids, glucagon-like peptide-1, and peptide-YY significantly changing postsurgery (P < .05 for all). Insulin sensitivity and insulin secretion rate during the MMTT significantly improved. Conclusion Three months post-VSG, youth showed significant improvements in weight, body composition, insulin sensitivity and secretion, and glycemic control, with most no longer requiring diabetes medications.
Collapse
Affiliation(s)
- Tyler J Dobbs
- University of Colorado Anschutz Medical Campus and Children's Hospital Colorado, Aurora, CO 80045, USA
| | - Melanie G Cree
- University of Colorado Anschutz Medical Campus and Children's Hospital Colorado, Aurora, CO 80045, USA
| | - Alex J Bailey
- University of Colorado Anschutz Medical Campus and Children's Hospital Colorado, Aurora, CO 80045, USA
| | - Amy D Baumgartner
- University of Colorado Anschutz Medical Campus and Children's Hospital Colorado, Aurora, CO 80045, USA
| | | | | | - Laura Pyle
- University of Colorado Anschutz Medical Campus and Children's Hospital Colorado, Aurora, CO 80045, USA
| | - Megan M Kelsey
- University of Colorado Anschutz Medical Campus and Children's Hospital Colorado, Aurora, CO 80045, USA
| | - Amy S Shah
- The University of Cincinnati and Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Thomas H Inge
- Northwestern University and Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Petter Bjornstad
- University of Colorado Anschutz Medical Campus and Children's Hospital Colorado, Aurora, CO 80045, USA
- University of Washington and Seattle Children's Hospital, Seattle, WA 98105, USA
| | - Kristen J Nadeau
- University of Colorado Anschutz Medical Campus and Children's Hospital Colorado, Aurora, CO 80045, USA
| |
Collapse
|
8
|
Zhang FS, Li HJ, Yu X, Song YP, Ren YF, Qian XZ, Liu JL, Li WX, Huang YR, Gao K. Global trends and hotspots of type 2 diabetes in children and adolescents: A bibliometric study and visualization analysis. World J Diabetes 2025; 16:96032. [PMID: 39817223 PMCID: PMC11718446 DOI: 10.4239/wjd.v16.i1.96032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 09/30/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Epidemiological surveys indicate an increasing incidence of type 2 diabetes mellitus (T2DM) among children and adolescents worldwide. Due to rapid disease progression, severe long-term cardiorenal complications, a lack of effective treatment strategies, and substantial socioeconomic burdens, it has become an urgent public health issue that requires management and resolution. Adolescent T2DM differs from adult T2DM. Despite a significant increase in our understanding of youth-onset T2DM over the past two decades, the related review and evidence-based content remain limited. AIM To visualize the hotspots and trends in pediatric and adolescent T2DM research and to forecast their future research themes. METHODS This study utilized the terms "children", "adolescents", and "type 2 diabetes", retrieving relevant articles published between 1983 and 2023 from three citation databases within the Web of Science Core Collection (SCI, SSCI, ESCI). Utilizing CiteSpace and VoSviewer software, we analyze and visually represent the annual output of literature, countries involved, and participating institutions. This allows us to predict trends in this research field. Our analysis encompasses co-cited authors, journal overlays, citation overlays, time-zone views, keyword analysis, and reference analysis, etc. RESULTS A total of 9210 articles were included, and the annual publication volume in this field showed a steady growth trend. The United States had the highest number of publications and the highest H-index. The United States also had the most research institutions and the strongest research capacity. The global hot journals were primarily diabetes professional journals but also included journals related to nutrition, endocrinology, and metabolism. Keyword analysis showed that research related to endothelial dysfunction, exposure risk, cardiac metabolic risk, changes in gut microbiota, the impact on comorbidities and outcomes, etc., were emerging keywords. They have maintained their popularity in this field, suggesting that these areas have garnered significant research interest in recent years. CONCLUSION Pediatric and adolescent T2DM is increasingly drawing global attention, with genes, behaviors, environmental factors, and multisystemic interventions potentially emerging as future research hot spots.
Collapse
Affiliation(s)
- Fang-Shuo Zhang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Hai-Jing Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xue Yu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yi-Ping Song
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yan-Feng Ren
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xuan-Zhu Qian
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jia-Li Liu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Wen-Xun Li
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yi-Ran Huang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Kuo Gao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| |
Collapse
|
9
|
Shah AS, Barrientos-Pérez M, Chang N, Fu JF, Hannon TS, Kelsey M, Peña AS, Pinhas-Hamiel O, Urakami T, Wicklow B, Wong J, Mahmud FH. ISPAD Clinical Practice Consensus Guidelines 2024: Type 2 Diabetes in Children and Adolescents. Horm Res Paediatr 2024; 97:555-583. [PMID: 39675348 PMCID: PMC11854986 DOI: 10.1159/000543033] [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: 11/01/2024] [Accepted: 11/23/2024] [Indexed: 12/17/2024] Open
Abstract
Youth-onset type 2 diabetes (T2D) results from genetic, environmental, and metabolic causes that differ among individuals and populations. This chapter builds on the 2022 ISPAD guidelines and summarizes recent advances in the management of T2D in children and adolescents. Updates include diagnostic algorithm for youth with new onset T2D, algorithms and tables for treatment, management, and assessment of comorbidities and complications and recommendations on recently approved pharmacologic therapies for the treatment of youth-onset T2D and management strategies. Youth-onset type 2 diabetes (T2D) results from genetic, environmental, and metabolic causes that differ among individuals and populations. This chapter builds on the 2022 ISPAD guidelines and summarizes recent advances in the management of T2D in children and adolescents. Updates include diagnostic algorithm for youth with new onset T2D, algorithms and tables for treatment, management, and assessment of comorbidities and complications and recommendations on recently approved pharmacologic therapies for the treatment of youth-onset T2D and management strategies.
Collapse
Affiliation(s)
- Amy S. Shah
- Division of Endocrinology, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati, Cincinnati, OH, USA
| | | | - Nancy Chang
- Center for Endocrinology, Diabetes and Metabolism, Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | - Jun-Fen Fu
- Department of Endocrinology, Children’s Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Tamara S. Hannon
- Division of Endocrinology, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Megan Kelsey
- Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, CO, USA
| | - Alexia S. Peña
- Robinson Research Institute and Women’s and Children’s Hospital, The University of Adelaide, North Adelaide, SA, Australia
| | - Orit Pinhas-Hamiel
- Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel-Hashomer, Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | | | - Brandy Wicklow
- Division of Endocrinology, Children’s Hospital Research Institute of Manitoba, Winnipeg Children’s Hospital and University of Manitoba, Winnipeg, MB, Canada
| | - Jencia Wong
- Department of Endocrinology, Royal Prince Alfred Hospital and Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Farid H. Mahmud
- Division of Endocrinology, Hospital for Sick Children, Sick Kids Research Institute, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
10
|
Bacha F, Hannon TS, Tosur M, Pike JM, Butler A, Tommerdahl KL, Zeitler PS. Pathophysiology and Treatment of Prediabetes and Type 2 Diabetes in Youth. Diabetes Care 2024; 47:2038-2049. [PMID: 39250166 PMCID: PMC11655414 DOI: 10.2337/dci24-0029] [Citation(s) in RCA: 5] [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: 03/15/2024] [Accepted: 07/20/2024] [Indexed: 09/10/2024]
Abstract
Youth-onset type 2 diabetes is a heterogeneous disease with increasing prevalence in relation to increased rates of obesity in children. It has genetic, epigenetic, social, and environmental determinants. Youth-onset type 2 diabetes is alarming given a rapidly progressive course compared with the course of adult-onset disease, early-onset vascular complications, and long-term exposure to hyperglycemia and associated complications. It is often preceded by prediabetes, a disease phase where defects in β-cell function relative to insulin sensitivity emerge. Herein, we review the current understanding of the pathophysiology of prediabetes and type 2 diabetes in youth. We describe the mechanisms underlying insulin resistance, the precipitous decline of β-cell function, and the role of other hormonal abnormalities in the pathogenesis of the disease. We discuss the critical importance of social determinants of health in the predisposition and progression of these conditions and present current management strategies and the advances in therapeutic approaches. These must adapt to meet the unique needs of the individual patient and family. Significant knowledge gaps remain that need to be addressed in future research.
Collapse
Affiliation(s)
- Fida Bacha
- USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX
- Division of Diabetes and Endocrinology, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX
| | - Tamara S. Hannon
- Division of Pediatric Endocrinology and Diabetology, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
- Pediatric Accelerator for Careers Engaged in Research, Children’s Health Services Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
| | - Mustafa Tosur
- USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX
- Division of Diabetes and Endocrinology, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX
| | - Julie M. Pike
- Division of Pediatric Endocrinology and Diabetology, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
- Pediatric Accelerator for Careers Engaged in Research, Children’s Health Services Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
| | - Ashley Butler
- Division of Psychology, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX
| | - Kalie L. Tommerdahl
- Section of Endocrinology, Department of Pediatrics, Children’s Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, CO
- Barbara Davis Center for Childhood Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO
- Ludeman Family Center for Women’s Health Research, Division of General Internal Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Philip S. Zeitler
- Section of Endocrinology, Department of Pediatrics, Children’s Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
11
|
Raithatha D, Sen P, Dave C, Yadav V, Mahapatra A, Shukla R, Bajpai A. Determinants of Course of Type 2 Diabetes in Indian Adolescents. Indian J Pediatr 2024; 91:1244-1249. [PMID: 38829540 DOI: 10.1007/s12098-024-05157-3] [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: 01/24/2024] [Accepted: 05/03/2024] [Indexed: 06/05/2024]
Abstract
OBJECTIVES To identify determinants of the course of Type 2 diabetes in Indian adolescents. METHODS Records of 37 adolescents (24 boys; 29 post pubertal and eight pubertal) with Type 2 diabetes (initial HbA1C 10.1 ± 1.9% and BMI SDS 2.0 ± 0.8; family history of diabetes in 33, 89.2%) diagnosed at 15.2 ± 2.5 y and followed up for 3.8 ± 2.2 y till 19.1 ± 3.3 y of age, were reviewed. RESULTS Initial treatment included insulin in 11 (29.7%), metformin alone in 22 (59.5%), and a combination of anti-diabetic medication in four (10.8%). Eleven subjects (29.7%) achieved remission at a median period of 5.3 mo (IQR- 17.13) after diagnosis; six of these relapsed within 0.9 ± 0.3 (range 0.4-1.3) y. The proportion of subjects requiring multiple anti-diabetic agents increased over follow-up (19% at six months, 32.5% at one year, 50% at two years, 59.1% at three, and 64.8% at four years), with the need for combination therapy after 0.9 ± 1.4 y. At the last follow-up, five were off treatment (13.5%), 10 (27%) were on metformin alone, and 22 (59.5%) were on multiple medications. The need for combination therapy at the last follow-up was lower in subjects with remission (27% against 73.1%, p = 0.02). CONCLUSIONS The findings of this study suggest delayed presentation and rapid progression of Type 2 diabetes in Indian adolescents. Diagnosis on screening and achievement of remission were predictors of good outcome.
Collapse
Affiliation(s)
- Dhvani Raithatha
- Department of Pediatric Endocrinology, Regency Center for Diabetes Endocrinology & Research, Regency City Clinic, Opposite PPN Market, Kanpur, 208001, India
| | - Proteek Sen
- Department of Pediatric Endocrinology, Regency Center for Diabetes Endocrinology & Research, Regency City Clinic, Opposite PPN Market, Kanpur, 208001, India
| | - Chetankumar Dave
- Department of Pediatric Endocrinology, Regency Center for Diabetes Endocrinology & Research, Regency City Clinic, Opposite PPN Market, Kanpur, 208001, India
| | - Vibha Yadav
- Department of Pediatric Endocrinology, Regency Center for Diabetes Endocrinology & Research, Regency City Clinic, Opposite PPN Market, Kanpur, 208001, India
| | - Alapan Mahapatra
- Department of Pediatric Endocrinology, Regency Center for Diabetes Endocrinology & Research, Regency City Clinic, Opposite PPN Market, Kanpur, 208001, India
| | - Rishi Shukla
- Department of Pediatric Endocrinology, Regency Center for Diabetes Endocrinology & Research, Regency City Clinic, Opposite PPN Market, Kanpur, 208001, India
| | - Anurag Bajpai
- Department of Pediatric Endocrinology, Regency Center for Diabetes Endocrinology & Research, Regency City Clinic, Opposite PPN Market, Kanpur, 208001, India.
| |
Collapse
|
12
|
Perry AS, Piaggi P, Huang S, Nayor M, Freedman J, North KE, Below JE, Clish CB, Murthy VL, Krakoff J, Shah RV. Human metabolic chambers reveal a coordinated metabolic-physiologic response to nutrition. JCI Insight 2024; 9:e184279. [PMID: 39576013 PMCID: PMC11601946 DOI: 10.1172/jci.insight.184279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 09/25/2024] [Indexed: 11/27/2024] Open
Abstract
Human studies linking metabolism with organism-wide physiologic function have been challenged by confounding, adherence, and precisionHere, we united physiologic and molecular phenotypes of metabolism during controlled dietary intervention to understand integrated metabolic-physiologic responses to nutrition. In an inpatient study of individuals who underwent serial 24-hour metabolic chamber experiments (indirect calorimetry) and metabolite profiling, we mapped a human metabolome onto substrate oxidation rates and energy expenditure across up to 7 dietary conditions (energy balance, fasting, multiple 200% caloric excess overfeeding of varying fat, protein, and carbohydrate composition). Diets exhibiting greater fat oxidation (e.g., fasting, high-fat) were associated with changes in metabolites within pathways of mitochondrial β-oxidation, ketogenesis, adipose tissue fatty acid liberation, and/or multiple anapleurotic substrates for tricarboxylic acid cycle flux, with inverse associations for diets with greater carbohydrate availability. Changes in each of these metabolite classes were strongly related to 24-hour respiratory quotient (RQ) and substrate oxidation rates (e.g., acylcarnitines related to lower 24-hour RQ and higher 24-hour lipid oxidation), underscoring links between substrate availability, physiology, and metabolism in humans. Physiologic responses to diet determined by gold-standard human metabolic chambers are strongly coordinated with biologically consistent, interconnected metabolic pathways encoded in the metabolome.
Collapse
Affiliation(s)
- Andrew S. Perry
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Paolo Piaggi
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Phoenix, Arizona, USA
| | - Shi Huang
- Department of Biostatistics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Matthew Nayor
- Sections of Cardiovascular Medicine and Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Jane Freedman
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Kari E. North
- CVD Genetic Epidemiology Computational Laboratory, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Jennifer E. Below
- Vanderbilt Genetics Institute, Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Clary B. Clish
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts USA
| | | | - Jonathan Krakoff
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Phoenix, Arizona, USA
| | - Ravi V. Shah
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
13
|
Abu-Nejem R, Hannon TS. Insulin Dynamics and Pathophysiology in Youth-Onset Type 2 Diabetes. J Clin Endocrinol Metab 2024; 109:2411-2421. [PMID: 38963882 DOI: 10.1210/clinem/dgae463] [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: 05/25/2024] [Revised: 06/28/2024] [Accepted: 07/03/2024] [Indexed: 07/06/2024]
Abstract
Youth-onset type 2 diabetes (T2D) is increasing around the globe. The mounting disease burden of youth-onset T2D portends substantial consequences for the health outcomes of young people and for health care systems. The pathophysiology of this condition is characterized by insulin resistance and initial insulin hypersecretion ± an inherent insulin secretory defect, with progressive loss of stimulated insulin secretion leading to pancreatic β-cell failure. Research studies focusing on youth-onset T2D have illuminated key differences for youth- vs adult-onset T2D, with youth having more profound insulin resistance and quicker progression to loss of sufficient insulin secretion to maintain euglycemia. There is a need for therapies that are targeted to improve both insulin resistance and, importantly, maintain sufficient insulin secretory function over the lifespan in youth-onset T2D.
Collapse
Affiliation(s)
- Rozan Abu-Nejem
- Department of Pediatrics, Divisions of Pediatric Endocrinology and Diabetology and Pediatric Health Services Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Tamara S Hannon
- Department of Pediatrics, Divisions of Pediatric Endocrinology and Diabetology and Pediatric Health Services Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
14
|
Abd-Elmoniem KZ, Edwan JH, Dietsche KB, Villalobos-Perez A, Shams N, Matta J, Baumgarten L, Qaddumi WN, Dixon SA, Chowdhury A, Stagliano M, Mabundo L, Wentzel A, Hadigan C, Gharib AM, Chung ST. Endothelial Dysfunction in Youth-Onset Type 2 Diabetes: A Clinical Translational Study. Circ Res 2024; 135:639-650. [PMID: 39069898 PMCID: PMC11361354 DOI: 10.1161/circresaha.124.324272] [Citation(s) in RCA: 3] [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: 01/22/2024] [Revised: 07/12/2024] [Accepted: 07/17/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND Youth-onset type 2 diabetes (Y-T2D) is associated with increased risk for coronary atherosclerotic disease, but the timing of the earliest pathological features and evidence of cardiac endothelial dysfunction have not been evaluated in this population. Endothelial function magnetic resonance imaging may detect early and direct endothelial dysfunction in the absence of classical risk factors (severe hyperglycemia, hypertension, and hyperlipidemia). Using endothelial function magnetic resonance imaging, we evaluated peripheral and coronary artery structure and endothelial function in young adults with Y-T2D diagnosed ≤5 years compared with age-matched healthy peers. We isolated and characterized plasma-derived small extracellular vesicles and evaluated their effects on inflammatory and signaling biomarkers in healthy human coronary artery endothelial cells to validate the imaging findings. METHODS Right coronary wall thickness, coronary artery flow-mediated dilation, and brachial artery flow-mediated dilation were measured at baseline and during isometric handgrip exercise using a 3.0T magnetic resonance imaging. Human coronary artery endothelial cells were treated with Y-T2D plasma-derived small extracellular vesicles. Protein expression was measured by Western blot analysis, oxidative stress was measured using the redox-sensitive probe dihydroethidium, and nitric oxide levels were measured by 4-amino-5-methylamino-2',7'-difluororescein diacetate. RESULTS Y-T2D (n=20) had higher hemoglobin A1c and high-sensitivity C-reactive protein, but similar total and LDL (low-density lipoprotein)-cholesterol compared with healthy peers (n=16). Y-T2D had greater coronary wall thickness (1.33±0.13 versus 1.22±0.13 mm; P=0.04) and impaired endothelial function: lower coronary artery flow-mediated dilation (-3.1±15.5 versus 15.9±17.3%; P<0.01) and brachial artery flow-mediated dilation (6.7±14.7 versus 26.4±15.2%; P=0.001). Y-T2D plasma-derived small extracellular vesicles reduced phosphorylated endothelial nitric oxide synthase expression and nitric oxide levels, increased reactive oxygen species production, and elevated ICAM (intercellular adhesion molecule)-mediated inflammatory pathways in human coronary artery endothelial cells. CONCLUSIONS Coronary and brachial endothelial dysfunction was evident in Y-T2D who were within 5 years of diagnosis and did not have severe hyperglycemia or dyslipidemia. Plasma-derived small extracellular vesicles induced markers of endothelial dysfunction, which corroborated accelerated subclinical coronary atherosclerosis as an early feature in Y-T2D. REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifier: NCT02830308 and NCT01399385.
Collapse
Affiliation(s)
- Khaled Z. Abd-Elmoniem
- National Institute of Diabetes and Digestive and Kidney Diseases, Biomedical Medical and Imaging Branch (K.Z.A., J.E., N.S., J.M., L.B., W.Q., A.M.G.), National Institutes of Health, Bethesda, MD
| | - Jehad H. Edwan
- National Institute of Diabetes and Digestive and Kidney Diseases, Biomedical Medical and Imaging Branch (K.Z.A., J.E., N.S., J.M., L.B., W.Q., A.M.G.), National Institutes of Health, Bethesda, MD
| | - Katrina B. Dietsche
- Diabetes Endocrinology and Obesity Branch (K.B., A.V., S.D., A.C., M.S., L.M., S.T.C.), National Institutes of Health, Bethesda, MD
| | - Alfredo Villalobos-Perez
- Diabetes Endocrinology and Obesity Branch (K.B., A.V., S.D., A.C., M.S., L.M., S.T.C.), National Institutes of Health, Bethesda, MD
| | - Nour Shams
- National Institute of Diabetes and Digestive and Kidney Diseases, Biomedical Medical and Imaging Branch (K.Z.A., J.E., N.S., J.M., L.B., W.Q., A.M.G.), National Institutes of Health, Bethesda, MD
| | - Jatin Matta
- National Institute of Diabetes and Digestive and Kidney Diseases, Biomedical Medical and Imaging Branch (K.Z.A., J.E., N.S., J.M., L.B., W.Q., A.M.G.), National Institutes of Health, Bethesda, MD
| | - Leilah Baumgarten
- National Institute of Diabetes and Digestive and Kidney Diseases, Biomedical Medical and Imaging Branch (K.Z.A., J.E., N.S., J.M., L.B., W.Q., A.M.G.), National Institutes of Health, Bethesda, MD
| | - Waleed N. Qaddumi
- National Institute of Diabetes and Digestive and Kidney Diseases, Biomedical Medical and Imaging Branch (K.Z.A., J.E., N.S., J.M., L.B., W.Q., A.M.G.), National Institutes of Health, Bethesda, MD
| | - Sydney A. Dixon
- Diabetes Endocrinology and Obesity Branch (K.B., A.V., S.D., A.C., M.S., L.M., S.T.C.), National Institutes of Health, Bethesda, MD
| | - Aruba Chowdhury
- Diabetes Endocrinology and Obesity Branch (K.B., A.V., S.D., A.C., M.S., L.M., S.T.C.), National Institutes of Health, Bethesda, MD
| | - Michael Stagliano
- Diabetes Endocrinology and Obesity Branch (K.B., A.V., S.D., A.C., M.S., L.M., S.T.C.), National Institutes of Health, Bethesda, MD
| | - Lilian Mabundo
- Diabetes Endocrinology and Obesity Branch (K.B., A.V., S.D., A.C., M.S., L.M., S.T.C.), National Institutes of Health, Bethesda, MD
| | - Annemarie Wentzel
- Hypertension in Africa Research Team (A.W.), North-West University, Potchefstroom
- South African Medical Research Council, Unit for Hypertension and Cardiovascular Disease (A.W.), North-West University, Potchefstroom
| | - Colleen Hadigan
- Clinical Center (C.H.), National Institutes of Health, Bethesda, MD
| | - Ahmed M. Gharib
- National Institute of Diabetes and Digestive and Kidney Diseases, Biomedical Medical and Imaging Branch (K.Z.A., J.E., N.S., J.M., L.B., W.Q., A.M.G.), National Institutes of Health, Bethesda, MD
| | - Stephanie T. Chung
- Diabetes Endocrinology and Obesity Branch (K.B., A.V., S.D., A.C., M.S., L.M., S.T.C.), National Institutes of Health, Bethesda, MD
| |
Collapse
|
15
|
Titmuss A, Korula S, Wicklow B, Nadeau KJ. Youth-onset Type 2 Diabetes: An Overview of Pathophysiology, Prognosis, Prevention and Management. Curr Diab Rep 2024; 24:183-195. [PMID: 38958831 PMCID: PMC11269415 DOI: 10.1007/s11892-024-01546-2] [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] [Accepted: 06/19/2024] [Indexed: 07/04/2024]
Abstract
PURPOSE OF REVIEW This review explores the emerging evidence regarding pathogenesis, future trajectories, treatment options, and phenotypes of youth-onset type 2 diabetes (T2D). RECENT FINDINGS Youth-onset T2D is increasing in incidence and prevalence worldwide, disproportionately affecting First Nations communities, socioeconomically disadvantaged youth, and people of colour. Youth-onset T2D differs in pathogenesis to later-onset T2D and progresses more rapidly. It is associated with more complications, and these occur earlier. While there are limited licensed treatment options available, the available medications also appear to have a poorer response in youth with T2D. Multiple interacting factors likely contribute to this rising prevalence, as well as the increased severity of the condition, including structural inequities, increasing obesity and sedentary lifestyles, and intergenerational transmission from in-utero exposure to maternal hyperglycemia and obesity. Youth-onset T2D is also associated with stigma and poorer mental health, and these impact clinical management. There is an urgent need to develop effective interventions to prevent youth-onset T2D and enhance engagement of affected youth. It is also critical to better understand the differing phenotypes of youth-onset T2D, to effectively target treatments, and to address intergenerational transmission in high-risk populations.
Collapse
Affiliation(s)
- Angela Titmuss
- Wellbeing and Preventable Chronic Diseases Division, Menzies School of Health Research, Charles Darwin University, Casuarina, PO Box 41096, Darwin, Northern Territory, Australia.
- Department of Paediatrics, Division of Women, Child and Youth, Royal Darwin Hospital, Darwin, Northern Territory, Australia.
| | - Sophy Korula
- Paediatric Endocrinology and Metabolism Division, Paediatric Unit-1, Christian Medical College Hospital, Vellore, India
- Department of Paediatrics, Latrobe Regional Hospital, Traralgon, Victoria, Australia
| | - Brandy Wicklow
- Department of Paediatrics and Child Health, University of Manitoba, Winnipeg, Canada
- Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Kristen J Nadeau
- Children's Hospital Colorado, Aurora, Colorado, USA
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
16
|
La Grasta Sabolic L, Marusic S, Cigrovski Berkovic M. Challenges and pitfalls of youth-onset type 2 diabetes. World J Diabetes 2024; 15:876-885. [PMID: 38766423 PMCID: PMC11099376 DOI: 10.4239/wjd.v15.i5.876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/04/2024] [Accepted: 04/01/2024] [Indexed: 05/10/2024] Open
Abstract
The incidence and prevalence of youth-onset type 2 diabetes mellitus (T2DM) are increasing. The rise in frequency and severity of childhood obesity, inclination to sedentary lifestyle, and epigenetic risks related to prenatal hyperglycemia exposure are important drivers of the youth-onset T2DM epidemic and might as well be responsible for the early onset of diabetes complications. Indeed, youth-onset T2DM has a more extreme metabolic phenotype than adult-onset T2DM, with greater insulin resistance and more rapid deterioration of beta cell function. Therefore, intermediate complications such as microalbuminuria develop in late childhood or early adulthood, while end-stage complications develop in mid-life. Due to the lack of efficacy and safety data, several drugs available for the treatment of adults with T2DM have not been approved in youth, reducing the pharmacological treatment options. In this mini review, we will try to address the present challenges and pitfalls related to youth-onset T2DM and summarize the available interventions to mitigate the risk of microvascular and macrovascular complications.
Collapse
Affiliation(s)
- Lavinia La Grasta Sabolic
- Department of Pediatric Endocrinology and Diabetology, University Hospital Centre Sestre Milosrdnice, Zagreb 10000, Croatia
- School of Medicine, Catholic University of Croatia, Zagreb 10000, Croatia
| | - Sanda Marusic
- Department for Sport and Exercise Medicine, University of Zagreb Faculty of Kinesiology , Zagreb 10000, Croatia
| | - Maja Cigrovski Berkovic
- Department for Sport and Exercise Medicine, University of Zagreb Faculty of Kinesiology , Zagreb 10000, Croatia
| |
Collapse
|
17
|
Dietsche KB, Magge SN, Dixon SA, Davis FS, Krenek A, Chowdhury A, Mabundo L, Stagliano M, Courville AB, Yang S, Turner S, Cai H, Kasturi K, Sherman AS, Ha J, Shouppe E, Walter M, Walter PJ, Chen KY, Brychta RJ, Peer C, Zeng Y, Figg W, Cogen F, Estrada DE, Chacko S, Chung ST. Glycemia and Gluconeogenesis With Metformin and Liraglutide: A Randomized Trial in Youth-onset Type 2 Diabetes. J Clin Endocrinol Metab 2024; 109:1361-1370. [PMID: 37967247 PMCID: PMC11031226 DOI: 10.1210/clinem/dgad669] [Citation(s) in RCA: 3] [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: 07/10/2023] [Revised: 11/02/2023] [Accepted: 11/13/2023] [Indexed: 11/17/2023]
Abstract
OBJECTIVE Elevated rates of gluconeogenesis are an early pathogenic feature of youth-onset type 2 diabetes (Y-T2D), but targeted first-line therapies are suboptimal, especially in African American (AA) youth. We evaluated glucose-lowering mechanisms of metformin and liraglutide by measuring rates of gluconeogenesis and β-cell function after therapy in AA Y-T2D. METHODS In this parallel randomized clinical trial, 22 youth with Y-T2D-age 15.3 ± 2.1 years (mean ± SD), 68% female, body mass index (BMI) 40.1 ± 7.9 kg/m2, duration of diagnosis 1.8 ± 1.3 years-were randomized to metformin alone (Met) or metformin + liraglutide (Lira) (Met + Lira) and evaluated before and after 12 weeks. Stable isotope tracers were used to measure gluconeogenesis [2H2O] and glucose production [6,6-2H2]glucose after an overnight fast and during a continuous meal. β-cell function (sigma) and whole-body insulin sensitivity (mSI) were assessed during a frequently sampled 2-hour oral glucose tolerance test. RESULTS At baseline, gluconeogenesis, glucose production, and fasting and 2-hour glucose were comparable in both groups, though Met + Lira had higher hemoglobin A1C. Met + Lira had a greater decrease from baseline in fasting glucose (-2.0 ± 1.3 vs -0.6 ± 0.9 mmol/L, P = .008) and a greater increase in sigma (0.72 ± 0.68 vs -0.05 ± 0.71, P = .03). The change in fractional gluconeogenesis was similar between groups (Met + Lira: -0.36 ± 9.4 vs Met: 0.04 ± 12.3%, P = .9), and there were no changes in prandial gluconeogenesis or mSI. Increased glucose clearance in both groups was related to sigma (r = 0.63, P = .003) but not gluconeogenesis or mSI. CONCLUSION Among Y-T2D, metformin with or without liraglutide improved glycemia but did not suppress high rates of gluconeogenesis. Novel therapies that will enhance β-cell function and target the elevated rates of gluconeogenesis in Y-T2D are needed.
Collapse
Affiliation(s)
- Katrina B Dietsche
- National Institute of Diabetes, Digestive and Kidney Diseases/National Institutes of Health, Bethesda, MD 20892, USA
| | - Sheela N Magge
- Division of Pediatric Endocrinology and Diabetes, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sydney A Dixon
- National Institute of Diabetes, Digestive and Kidney Diseases/National Institutes of Health, Bethesda, MD 20892, USA
| | - Faith S Davis
- National Institute of Diabetes, Digestive and Kidney Diseases/National Institutes of Health, Bethesda, MD 20892, USA
| | - Andrea Krenek
- National Institute of Diabetes, Digestive and Kidney Diseases/National Institutes of Health, Bethesda, MD 20892, USA
| | - Aruba Chowdhury
- National Institute of Diabetes, Digestive and Kidney Diseases/National Institutes of Health, Bethesda, MD 20892, USA
| | - Lilian Mabundo
- National Institute of Diabetes, Digestive and Kidney Diseases/National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael Stagliano
- National Institute of Diabetes, Digestive and Kidney Diseases/National Institutes of Health, Bethesda, MD 20892, USA
| | - Amber B Courville
- National Institute of Diabetes, Digestive and Kidney Diseases/National Institutes of Health, Bethesda, MD 20892, USA
| | - Shanna Yang
- Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sara Turner
- Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hongyi Cai
- National Institute of Diabetes, Digestive and Kidney Diseases/National Institutes of Health, Bethesda, MD 20892, USA
| | - Kannan Kasturi
- Division of Pediatric Endocrinology, Essentia Health, Duluth, MN 55805, USA
| | - Arthur S Sherman
- National Institute of Diabetes, Digestive and Kidney Diseases/National Institutes of Health, Bethesda, MD 20892, USA
| | - Joon Ha
- Department of Mathematics, Howard University, Washington, DC 20059, USA
| | - Eileen Shouppe
- National Institute of Diabetes, Digestive and Kidney Diseases/National Institutes of Health, Bethesda, MD 20892, USA
| | - Mary Walter
- National Institute of Diabetes, Digestive and Kidney Diseases/National Institutes of Health, Bethesda, MD 20892, USA
| | - Peter J Walter
- National Institute of Diabetes, Digestive and Kidney Diseases/National Institutes of Health, Bethesda, MD 20892, USA
| | - Kong Y Chen
- National Institute of Diabetes, Digestive and Kidney Diseases/National Institutes of Health, Bethesda, MD 20892, USA
| | - Robert J Brychta
- National Institute of Diabetes, Digestive and Kidney Diseases/National Institutes of Health, Bethesda, MD 20892, USA
| | - Cody Peer
- Clinical Pharmacology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yi Zeng
- Clinical Pharmacology Laboratory, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - William Figg
- Clinical Pharmacology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Fran Cogen
- Division of Endocrinology and Diabetes, Children's National Hospital, Washington, DC 20010, USA
| | - D Elizabeth Estrada
- Division of Endocrinology and Diabetes, Children's National Hospital, Washington, DC 20010, USA
| | - Shaji Chacko
- Department of Pediatrics, Children's Nutrition Research Center and Division of Pediatric Endocrinology and Metabolism, U.S. Department of Agriculture/Agricultural Research Service, Baylor College of Medicine, Houston, TX 77030, USA
| | - Stephanie T Chung
- National Institute of Diabetes, Digestive and Kidney Diseases/National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
18
|
Perry AS, Piaggi P, Huang S, Nayor M, Freedman J, North K, Below J, Clish C, Murthy VL, Krakoff J, Shah RV. Human metabolic chambers reveal a coordinated metabolic-physiologic response to nutrition. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.08.24305087. [PMID: 38645000 PMCID: PMC11030300 DOI: 10.1101/2024.04.08.24305087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The emerging field of precision nutrition is based on the notion that inter-individual responses across diets of different calorie-macronutrient content may contribute to inter-individual differences in metabolism, adiposity, and weight gain. Free-living diet studies have been traditionally challenged by difficulties in controlling adherence to prescribed calories and macronutrient content and rarely allow a period of metabolic stability prior to metabolic measures (to minimize influences of weight changes). In this context, key physiologic measures central to precision nutrition responses may be most precisely quantified via whole room indirect calorimetry over 24-h, in which precise control of activity and nutrition can be achieved. In addition, these studies represent unique "N of 1" human crossover metabolic-physiologic experiments during which specific molecular pathways central to nutrient metabolism may be discerned. Here, we quantified 263 circulating metabolites during a ≈40-day inpatient admission in which up to 94 participants underwent seven monitored 24-h nutritional interventions of differing macronutrient composition in a whole-room indirect calorimeter to capture precision metabolic responses. Broadly, we observed heterogenous responses in metabolites across dietary chambers, with the exception of carnitines which tracked with 24-h respiratory quotient. We identified excursions in shared metabolic species (e.g., carnitines, glycerophospholipids, amino acids) that mapped onto gold-standard calorimetric measures of substrate oxidation preference and lipid availability. These findings support a coordinated metabolic-physiologic response to nutrition, highlighting the relevance of these controlled settings to uncover biological pathways of energy utilization during precision nutrition studies.
Collapse
|
19
|
Ha J, Chung ST, Springer M, Kim JY, Chen P, Chhabra A, Cree MG, Diniz Behn C, Sumner AE, Arslanian SA, Sherman AS. Estimating insulin sensitivity and β-cell function from the oral glucose tolerance test: validation of a new insulin sensitivity and secretion (ISS) model. Am J Physiol Endocrinol Metab 2024; 326:E454-E471. [PMID: 38054972 PMCID: PMC11639675 DOI: 10.1152/ajpendo.00189.2023] [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: 06/23/2023] [Revised: 11/27/2023] [Accepted: 11/27/2023] [Indexed: 12/07/2023]
Abstract
Efficient and accurate methods to estimate insulin sensitivity (SI) and β-cell function (BCF) are of great importance for studying the pathogenesis and treatment effectiveness of type 2 diabetes (T2D). Existing methods range in sensitivity, input data, and technical requirements. Oral glucose tolerance tests (OGTTs) are preferred because they are simpler and more physiological than intravenous methods. However, current analytical methods for OGTT-derived SI and BCF also range in complexity; the oral minimal models require mathematical expertise for deconvolution and fitting differential equations, and simple algebraic surrogate indices (e.g., Matsuda index, insulinogenic index) may produce unphysiological values. We developed a new insulin secretion and sensitivity (ISS) model for clinical research that provides precise and accurate estimates of SI and BCF from a standard OGTT, focusing on effectiveness, ease of implementation, and pragmatism. This model was developed by fitting a pair of differential equations to glucose and insulin without need of deconvolution or C-peptide data. This model is derived from a published model for longitudinal simulation of T2D progression that represents glucose-insulin homeostasis, including postchallenge suppression of hepatic glucose production and first- and second-phase insulin secretion. The ISS model was evaluated in three diverse cohorts across the lifespan. The new model had a strong correlation with gold-standard estimates from intravenous glucose tolerance tests and insulin clamps. The ISS model has broad applicability among diverse populations because it balances performance, fidelity, and complexity to provide a reliable phenotype of T2D risk.NEW & NOTEWORTHY The pathogenesis of type 2 diabetes (T2D) is determined by a balance between insulin sensitivity (SI) and β-cell function (BCF), which can be determined by gold standard direct measurements or estimated by fitting differential equation models to oral glucose tolerance tests (OGTTs). We propose and validate a new differential equation model that is simpler to use than current models and requires less data while maintaining good correlation and agreement with gold standards. Matlab and Python code is freely available.
Collapse
Affiliation(s)
- Joon Ha
- Department of Mathematics, Howard University, Washington, District of Columbia, United States
| | - Stephanie T Chung
- Section on Pediatric Diabetes, Obesity, and Metabolism, Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States
| | - Max Springer
- Department of Mathematics, University of Maryland, College Park, Maryland, United States
| | - Joon Young Kim
- Department of Exercise Science, David B. Falk College of Sport and Human Dynamics, Syracuse University, Syracuse, New York, United States
| | | | - Aaryan Chhabra
- Department of Biology, Indian Institute of Science Education and Research, Pune, India
| | - Melanie G Cree
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Cecilia Diniz Behn
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Department of Applied Mathematics and Statistics, Colorado School of Mines, Golden, Colorado, United States
| | - Anne E Sumner
- Intramural Research Program, National Institute on Minority Health and Health Disparities (NIMHD), National Institutes of Health, Bethesda, Maryland, United States
- Section on Ethnicity and Health, Diabetes Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, Maryland, United States
- Hypertension in Africa Research Team, North-West University, Potchefstroom, South Africa
| | - Silva A Arslanian
- Division of Pediatric Endocrinology, Metabolism and Diabetes Mellitus, Center for Pediatric Research in Obesity and Metabolism, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Arthur S Sherman
- Laboratory of Biological Modeling, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
20
|
Bacha F, El-Ayash H, Mohamad M, Sharma S, Puyau M, Kanchi R, Coarfa C. Distinct Amino Acid Profile Characterizes Youth With or at Risk for Type 2 Diabetes. Diabetes 2024; 73:628-636. [PMID: 38215171 DOI: 10.2337/db23-0375] [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: 05/12/2023] [Accepted: 01/10/2024] [Indexed: 01/14/2024]
Abstract
Branched-chain amino acids (BCAAs) and aromatic AAs (AAAs) are associated with increased risk for type 2 diabetes in adults. Studies in youth show conflicting results. We hypothesized that an AA metabolomic signature can be defined to identify youth at risk for β-cell failure and the development of type 2 diabetes. We performed targeted AA metabolomics analysis on 127 adolescents (65 girls; 15.5 [SD ±1.9] years old, Tanner stage II-V) with normal weight or obesity across the spectrum of glycemia, with assessment of AA concentrations by mass spectrometry, at fasting, and steady state of a hyperinsulinemic-euglycemic clamp, with determination of insulin sensitivity (IS) per fat-free mass (FFM). We measured insulin secretion during a 2-h hyperglycemic clamp and calculated the disposition index per FFM (DIFFM), a measure of β-cell function. Our results showed that concentration of glycine (Gly) and the glutamine (Gln)-to-glutamate (Glu) ratio were lower, whereas BCAA, tyrosine, and lysine (Lys) concentrations were higher in the groups with obesity and dysglycemia compared with those with normal weight. Gly and Gln-to-Glu ratio were positively related to IS and DIFFM, with opposite relationships observed for BCAAs, AAAs, and Lys. We conclude that a metabolic signature of low Gly concentration and low Gln-to-Glu ratio, and elevated BCAAs, AAAs, and Lys concentrations may constitute a biomarker to identify youth at risk for β-cell failure. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Fida Bacha
- U.S. Department of Agriculture, Agricultural Research Service, Children's Nutrition Research Center, Houston, TX
- Division of Pediatric Endocrinology and Diabetes, Texas Children's Hospital, Houston, TX
- Baylor College of Medicine, Houston, TX
| | - Heba El-Ayash
- U.S. Department of Agriculture, Agricultural Research Service, Children's Nutrition Research Center, Houston, TX
- Division of Pediatric Endocrinology and Diabetes, Texas Children's Hospital, Houston, TX
- Baylor College of Medicine, Houston, TX
| | - Mahmoud Mohamad
- U.S. Department of Agriculture, Agricultural Research Service, Children's Nutrition Research Center, Houston, TX
- Baylor College of Medicine, Houston, TX
| | - Susan Sharma
- U.S. Department of Agriculture, Agricultural Research Service, Children's Nutrition Research Center, Houston, TX
- Baylor College of Medicine, Houston, TX
| | - Maurice Puyau
- U.S. Department of Agriculture, Agricultural Research Service, Children's Nutrition Research Center, Houston, TX
- Baylor College of Medicine, Houston, TX
| | | | | |
Collapse
|
21
|
Bjornstad P, Choi YJ, Platnick C, Gross S, Narongkiatikhun P, Melena I, Remmers L, Baca M, Schutte G, Dobbs T, Vigers T, Pyle L, Driscoll L, Tommerdahl K, Kendrick J, Looker HC, Dart A, Cherney D, van Raalte DH, Srivastava A, Li L, Prasad P, Saulnier P, Nelson RG, Johnson RJ, Nadeau KJ. Insulin Secretion, Sensitivity, and Kidney Function in Young Individuals With Type 2 Diabetes. Diabetes Care 2024; 47:409-417. [PMID: 38153805 PMCID: PMC10909687 DOI: 10.2337/dc23-1818] [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: 09/27/2023] [Accepted: 12/03/2023] [Indexed: 12/30/2023]
Abstract
OBJECTIVE β-Cell dysfunction and insulin resistance magnify the risk of kidney injury in type 2 diabetes. The relationship between these factors and intraglomerular hemodynamics and kidney oxygen availability in youth with type 2 diabetes remains incompletely explored. RESEARCH DESIGN AND METHODS Fifty youth with type 2 diabetes (mean age ± SD 16 ± 2 years; diabetes duration 2.3 ± 1.8 years; 60% female; median HbA1c 6.4% [25th, 75th percentiles 5.9, 7.6%]; BMI 36.4 ± 7.4 kg/m2; urine albumin-to-creatinine ratio [UACR] 10.3 [5.9, 58.0] mg/g) 21 control participants with obesity (OCs; age 16 ± 2 years; 29% female; BMI 37.6 ± 7.4 kg/m2), and 20 control participants in the normal weight category (NWCs; age 17 ± 3 years; 70% female; BMI 22.5 ± 3.6 kg/m2) underwent iohexol and p-aminohippurate clearance to assess glomerular filtration rate (GFR) and renal plasma flow, kidney MRI for oxygenation, hyperglycemic clamp for insulin secretion (acute C-peptide response to glucose [ACPRg]) and disposition index (DI; ×103 mg/kg lean/min), and DXA for body composition. RESULTS Youth with type 2 diabetes exhibited lower DI (0.6 [0.0, 1.6] vs. 3.8 [2.4, 4.5] × 103 mg/kg lean/min; P < 0.0001) and ACPRg (0.6 [0.3, 1.4] vs. 5.3 [4.3, 6.9] nmol/L; P < 0.001) and higher UACR (10.3 [5.9, 58.0] vs. 5.3 [3.4, 14.3] mg/g; P = 0.003) and intraglomerular pressure (77.8 ± 11.5 vs. 64.8 ± 5.0 mmHg; P < 0.001) compared with OCs. Youth with type 2 diabetes and OCs had higher GFR and kidney oxygen availability (relative hyperoxia) than NWCs. DI was associated inversely with intraglomerular pressure and kidney hyperoxia. CONCLUSIONS Youth with type 2 diabetes demonstrated severe β-cell dysfunction that was associated with intraglomerular hypertension and kidney hyperoxia. Similar but attenuated findings were found in OCs.
Collapse
Affiliation(s)
- Petter Bjornstad
- Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado School of Medicine, Aurora, CO
| | - Ye Ji Choi
- Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
- Department of Biostatistics and Informatics, University of Colorado School of Medicine, Aurora, CO
| | - Carson Platnick
- Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Susan Gross
- Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Phoom Narongkiatikhun
- Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Isabella Melena
- Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Lauryn Remmers
- Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Madison Baca
- Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Grant Schutte
- Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Tyler Dobbs
- Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Tim Vigers
- Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
- Department of Biostatistics and Informatics, University of Colorado School of Medicine, Aurora, CO
| | - Laura Pyle
- Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
- Department of Biostatistics and Informatics, University of Colorado School of Medicine, Aurora, CO
| | - Lynette Driscoll
- Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Kalie Tommerdahl
- Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Jessica Kendrick
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado School of Medicine, Aurora, CO
| | - Helen C. Looker
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ
| | - Allison Dart
- Division of Nephrology, Department of Pediatrics, University of Manitoba, Winnipeg, Manitoba, Canada
- Children’s Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - David Cherney
- Division of Nephrology, Department of Medicine, University of Toronto School of Medicine, Toronto, Ontario, Canada
| | - Daniel H. van Raalte
- Diabetes Center, Department of Internal Medicine, Vrije Universiteit University Medical Center, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Anand Srivastava
- Division of Nephrology, Department of Medicine, University of Illinois Chicago, Chicago, IL
| | - Luping Li
- Department of Radiology, NorthShore University HealthSystem, Evanston, IL
| | - Pottumarthi Prasad
- Department of Radiology, NorthShore University HealthSystem, Evanston, IL
| | - Pierre Saulnier
- INSERM Centre d’Investigation Clinique 1402, CHU Poitiers, University of Poitiers, Poitiers, France
| | - Robert G. Nelson
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ
| | - Richard J. Johnson
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado School of Medicine, Aurora, CO
| | - Kristen J. Nadeau
- Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| |
Collapse
|
22
|
Burton JJN, Alonso LC. Overnutrition in the early postnatal period influences lifetime metabolic risk: Evidence for impact on pancreatic β-cell mass and function. J Diabetes Investig 2024; 15:263-274. [PMID: 38193815 PMCID: PMC10906026 DOI: 10.1111/jdi.14136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 12/05/2023] [Indexed: 01/10/2024] Open
Abstract
Overconsumption of energy-rich foods that disrupt caloric balance is a fundamental cause of overweight, obesity and diabetes. Dysglycemia and the resulting cardiovascular disease cause substantial morbidity and mortality worldwide, as well as high societal cost. The prevalence of obesity in childhood and adolescence is increasing, leading to younger diabetes diagnosis, and higher severity of microvascular and macrovascular complications. An important goal is to identify early life conditions that increase future metabolic risk, toward the goal of preventing diabetes and cardiovascular disease. An ample body of evidence implicates prenatal and postnatal childhood growth trajectories in the programming of adult metabolic disease. Human epidemiological data show that accelerated childhood growth increases risk of type 2 diabetes in adulthood. Type 2 diabetes results from the combination of insulin resistance and pancreatic β-cell failure, but specific mechanisms by which accelerated postnatal growth impact one or both of these processes remain uncertain. This review explores the metabolic impact of overnutrition during postnatal life in humans and in rodent models, with specific attention to the connection between accelerated childhood growth and future adiposity, insulin resistance, β-cell mass and β-cell dysfunction. With improved knowledge in this area, we might one day be able to modulate nutrition and growth in the critical postnatal window to maximize lifelong metabolic health.
Collapse
Affiliation(s)
- Joshua JN Burton
- Division of Endocrinology, Diabetes and Metabolism and the Joan and Sanford I. Weill Center for Metabolic Health, Weill Cornell MedicineNew York CityNew YorkUSA
| | - Laura C Alonso
- Division of Endocrinology, Diabetes and Metabolism and the Joan and Sanford I. Weill Center for Metabolic Health, Weill Cornell MedicineNew York CityNew YorkUSA
| |
Collapse
|
23
|
Hua J, Lin H, Wang X, Qian ZM, Vaughn MG, Tabet M, Wang C, Lin H. Associations of glycosylated hemoglobin, pre-diabetes, and type 2 diabetes with incident lung cancer: A large prospective cohort study. Diabetes Metab Syndr 2024; 18:102968. [PMID: 38402819 DOI: 10.1016/j.dsx.2024.102968] [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: 10/07/2023] [Revised: 02/12/2024] [Accepted: 02/15/2024] [Indexed: 02/27/2024]
Abstract
BACKGROUND The association of pre-diabetes and type 2 diabetes (T2D) with incident lung cancer is uncertain, and the incident risk across the glycemic spectrum is unclear. We aimed to explore the associations of glycosylated hemoglobin (HbA1c), pre-diabetes, and T2D with incident lung cancer in a large prospective cohort. METHODS Leveraging a total of 210,779 cancer-free adults recruited in the UK Biobank between 2006 and 2010. We performed multivariable Cox proportional hazards models and restricted cubic spline methods to estimate hazard ratios (HRs) and 95% confidence intervals (95% CIs) for the associations of HbA1c, pre-diabetes, and T2D with incident lung cancer. RESULTS During a median follow-up of 11.06 years, 1738 incident lung cancer cases were ascertained. The incidence of lung cancer was 20% higher among people with diabetes (HR: 1.20, 95% CI: 1.02 to 1.42) and 38% higher among people with pre-diabetes (HR: 1.38, 95% CI: 1.15 to 1.65). After dividing people with diabetes by whether taking antidiabetic medications, the incidence was 28% higher among people with diabetes without medications (HR: 1.28, 95% CI: 1.02 to 1.61) and 15% higher among people with diabetes with medications (HR: 1.15, 95% CI: 0.93 to 1.41). The increased risk of incident lung cancer for each standard deviation (6.45 mmol/mol) increase in HbA1c was more pronounced across HbA1c values of 32-42 mmol/mol (HR: 1.37, 95% CI: 1.18 to 1.59). The risk was more pronounced among participants <60 years. CONCLUSIONS Pre-diabetes and T2D are associated with an increased incidence of lung cancer. The increased risk of incident lung cancer is more pronounced across HbA1c values of 32-42 mmol/mol, which are currently considered normal values.
Collapse
Affiliation(s)
- Junjie Hua
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Huan Lin
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Xiaojie Wang
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zhengmin Min Qian
- Department of Epidemiology and Biostatistics, College for Public Health & Social Justice, Saint Louis University, Saint Louis, 63104, USA
| | - Michael G Vaughn
- School of Social Work, Saint Louis University, St. Louis, 63103, USA
| | - Maya Tabet
- College of Global Population Health, University of Health Sciences and Pharmacy in St. Louis, St. Louis, 63110, USA
| | - Chongjian Wang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Hualiang Lin
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
24
|
Chung ST, Davis F, Patel T, Mabundo L, Estrada DE. Reevaluating First-line Therapies in Youth-Onset Type 2 Diabetes. J Clin Endocrinol Metab 2024; 109:e870-e872. [PMID: 37624230 PMCID: PMC11032239 DOI: 10.1210/clinem/dgad508] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/09/2023] [Accepted: 08/23/2023] [Indexed: 08/26/2023]
Abstract
The prevalence of youth-onset type 2 diabetes is growing worldwide and current first-line treatment with metformin and intensive behavior and lifestyle changes are suboptimal in over 50% of youth within 2 years of diagnosis. This perspective article is a call to action for reevaluation of existing strategies and critical appraisal of metformin as first-line therapy in youth-onset type 2 diabetes. Increased attention should be given to novel therapeutics approved in youth, including glucagon-like 1 receptor agonists, sodium glucose cotransporter-2, and sociocultural interventions that will promote diabetes self-management.
Collapse
Affiliation(s)
- Stephanie T Chung
- Section on Pediatric Diabetes, Obesity, and Metabolism, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
- Division of Endocrinology and Diabetes, Children’s National Hospital, Washington, DC 20010, USA
| | - Faith Davis
- Section on Pediatric Diabetes, Obesity, and Metabolism, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tejal Patel
- Division of Endocrinology and Diabetes, Children’s National Hospital, Washington, DC 20010, USA
| | - Lilian Mabundo
- Section on Pediatric Diabetes, Obesity, and Metabolism, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Doris E Estrada
- Division of Endocrinology and Diabetes, Children’s National Hospital, Washington, DC 20010, USA
| |
Collapse
|
25
|
Hitt TA, Hannon TS, Magge SN. Approach to the Patient: Youth-Onset Type 2 Diabetes. J Clin Endocrinol Metab 2023; 109:245-255. [PMID: 37584397 DOI: 10.1210/clinem/dgad482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/10/2023] [Accepted: 08/11/2023] [Indexed: 08/17/2023]
Abstract
Youth-onset type 2 diabetes is a growing epidemic with a rising incidence worldwide. Although the pathogenesis and diagnosis of youth-onset type 2 diabetes are similar to adult-onset type 2 diabetes, youth-onset type 2 diabetes is unique, with greater insulin resistance, insulin hypersecretion, and faster progression of pancreatic beta cell function decline. Individuals with youth-onset type 2 diabetes also develop complications at higher rates within short periods of time compared to adults with type 2 diabetes or youth with type 1 diabetes. The highest prevalence and incidence of youth-onset type 2 diabetes in the United States is among youth from minoritized racial and ethnic groups. Risk factors include obesity, family history of type 2 diabetes, comorbid conditions and use of medications associated with insulin resistance and rapid weight gain, socioeconomic and environmental stressors, and birth history of small-for-gestational-age or pregnancy associated with gestational or pregestational diabetes. Patients with youth-onset type 2 diabetes should be treated using a multidisciplinary model with frequent clinic visits and emphasis on addressing of social and psychological barriers to care and glycemic control, as well as close monitoring for comorbidities and complications. Intensive health behavior therapy is an important component of treatment, in addition to medical management, both of which should be initiated at the diagnosis of type 2 diabetes. There are limited but growing pharmacologic treatment options, including metformin, insulin, glucagon-like peptide 1 receptor agonists, and sodium-glucose cotransporter 2 inhibitors. Although long-term outcomes are not fully known, metabolic/bariatric surgery in youth with type 2 diabetes has led to improved cardiometabolic outcomes.
Collapse
Affiliation(s)
- Talia A Hitt
- Division of Endocrinology and Diabetes, Department of Pediatrics, Johns Hopkins University School of Medicine, 200 N. Wolfe Street, Room 3114, Baltimore, MD 21287, USA
| | - Tamara S Hannon
- Division of Endocrinology and Diabetology, Department of Pediatrics, Indiana University School of Medicine, 705 Riley Hospital Drive, Indianapolis, IN 46202, USA
| | - Sheela N Magge
- Division of Endocrinology and Diabetes, Department of Pediatrics, Johns Hopkins University School of Medicine, 200 N. Wolfe Street, Room 3114, Baltimore, MD 21287, USA
| |
Collapse
|
26
|
Ravà L, Fintini D, Mariani M, Deodati A, Inzaghi E, Pedicelli S, Bizzarri C, Cappa M, Cianfarani S, Manco M. High 1-h glucose in youths with obesity as marker of prediabetes and cardiovascular risk. J Endocrinol Invest 2023; 46:2555-2562. [PMID: 37204691 DOI: 10.1007/s40618-023-02111-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 05/09/2023] [Indexed: 05/20/2023]
Abstract
PURPOSE Testing 1-h glucose (1HG) concentration during oral glucose tolerance test is cost-effective to identify individuals at risk of incident type 2 diabetes. Aim of the study was to define 1HG cutoffs diagnostic of incident impaired glucose tolerance (IGT) in youths with obesity, and to evaluate prevalence and association of cutoffs identified in the cohort and from the literature (133 and 155 mg/dl) to cardiovascular disease (CVD) in a population of youths with obesity. METHODS This is a longitudinal study of 154 youths to identify 1HG cutoffs, and cross-sectional study of 2295 youths to estimate prevalence of high 1HG and association to CVD. Receiver-operating characteristic curves (ROC) were used to establish 1HG cutoffs, and univariate regression analyses to test association of 1HG to blood pressure, lipids and aminotransferases. RESULTS ROC analysis identified the 1HG cutoff of 159 mg/dl as having diagnostic accuracy of IGT with area under the ROC 0.82 (95% CI 0.66-0.98), sensitivity 0.86% and specificity 0.79%. In the cross-sectional population, prevalence of high 1HG was 36% and 15% for 133 and 155 mg/dl cutoffs, respectively, and 17% for the 159 mg/dl value. All the examined cutoffs were significantly associated with worse lipid profile, liver function test, reduced insulin sensitivity, secretion and disposition index. CONCLUSION High 1HG is marker of persistent IGT and increased risk of metabolic abnormalities in youths. The 155 mg/dl cutoff is a convenient estimate in young people but longitudinal studies with retinopathy and overt diabetes as end points are advised to verify the 1HG cutoff with the best diagnostic accuracy.
Collapse
Affiliation(s)
- L Ravà
- Clinical Epidemiology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - D Fintini
- Unit of Endocrinology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - M Mariani
- Unit of Endocrinology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - A Deodati
- Unit of Endocrinology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - E Inzaghi
- Unit of Endocrinology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - S Pedicelli
- Unit of Endocrinology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - C Bizzarri
- Unit of Endocrinology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - M Cappa
- Unit of Endocrinology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - S Cianfarani
- Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy
- Diabetes and Growth Disorders Unit, Bambino Gesù' Children's Hospital, IRCCS, Rome, Italy
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - M Manco
- Research Area for Multifactorial Diseases and Complex Phenotypes, Bambino Gesù Children's Hospital, IRCCS, Via F. Baldelli 38, 00146, Rome, Italy.
| |
Collapse
|
27
|
Fan Y, Fan B, Lau ESH, Lim CKP, Wu H, Ma RCW, Ozaki R, Kong APS, Chow E, Luk AOY, Chan JCN. Comparison of beta-cell function between Hong Kong Chinese with young-onset type 2 diabetes and late-onset type 2 diabetes. Diabetes Res Clin Pract 2023; 205:110954. [PMID: 37839755 DOI: 10.1016/j.diabres.2023.110954] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/09/2023] [Accepted: 10/13/2023] [Indexed: 10/17/2023]
Abstract
AIMS We compared beta-cell function in Chinese with type 2 diabetes diagnosed at age < 40 years (young-onset diabetes, YOD) and ≥ 40 years (late-onset diabetes, LOD). METHODS In this cross-sectional study, we selected participants from two cohorts of people with type 2 diabetes recruited in 1996-2012 (n = 4,376) and 2020-2021 (n = 794). Multivariable linear regression models were applied to compare homeostasis model assessment of beta-cell function (HOMA2-%B) and fasting plasma C-peptide across diabetes duration at enrolment between YOD and LOD. RESULTS The YOD group (n = 1,876, mean [SD] age: 39.9 [7.5] years, median [IQR] diabetes duration: 6 [2-12] years) was more likely to have family history of diabetes (61.6 % vs 43.6 %), obesity (41.9 % vs 26.8 %), dyslipidaemia (61.7 % vs 54.4 %), and worse glycaemic control (mean HbA1c 7.7 % vs 7.4 %) than those with LOD (n = 3,294, age: 60.8 [10.6] years, diabetes duration: 5 [1-10] years). When compared to people with LOD, HOMA2-%B and fasting plasma C-peptide were lower in the YOD group, consistently among those with BMI < 27.5 kg/m2 and HOMA2-IR ≤ 1.6 (median value), adjusted for year at enrolment, sex, diabetes duration, family history of diabetes, HbA1c, weight and lipid indices (p < 0.01). Cross-sectionally, the slopes of decline in HOMA2-%B by diabetes duration were greater in YOD than LOD among individuals with BMI < 27.5 kg/m2 (p-interaction = 0.015). CONCLUSIONS Chinese with YOD had accelerated loss of beta-cell function than those with LOD especially in non-obese individuals.
Collapse
Affiliation(s)
- Yingnan Fan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Baoqi Fan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region; Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Eric S H Lau
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Cadmon K P Lim
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Hongjiang Wu
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Ronald C W Ma
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region; Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong Special Administrative Region; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Risa Ozaki
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region; Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Alice P S Kong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region; Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong Special Administrative Region; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Elaine Chow
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region; Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Andrea O Y Luk
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region; Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong Special Administrative Region; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region.
| | - Juliana C N Chan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region; Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong Special Administrative Region; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| |
Collapse
|
28
|
Tjaden AH, Edelstein SL, Arslanian S, Barengolts E, Caprio S, Cree-Green M, Lteif A, Mather KJ, Savoye M, Xiang AH, Kahn SE. Reproducibility of Glycemic Measures Among Dysglycemic Youth and Adults in the RISE Study. J Clin Endocrinol Metab 2023; 108:e1125-e1133. [PMID: 36938582 PMCID: PMC10505524 DOI: 10.1210/clinem/dgad135] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 02/16/2023] [Accepted: 03/16/2023] [Indexed: 03/21/2023]
Abstract
AIMS Previous work found poor reproducibility for measures of glycemia in individuals at risk for dysglycemia. Differences between youth and adults have not been assessed. Using youth and adults in the Restoring Insulin Secretion Study, we tested variability and classification concordance for hemoglobin A1C (HbA1c), fasting and 2-hour glucose from oral glucose tolerance tests (OGTTs). METHODS HbA1c and glucose on repeated samples obtained ∼6 weeks apart were compared in 66 youth (mean age 14.2 years) and 354 adults (52.7 years). Changes, coefficient of variation (CV), and concordance of diagnostic categories between the 2 visits were compared. RESULTS Mean difference between the 2 visits in HbA1c was higher in youth than adults (P < .001), while fasting glucose was similar and 2-hour glucose was lower in youth (P = .051). CV was smallest for HbA1c compared to fasting and 2-hour glucose. For HbA1c, youth had higher CV (P < .001); whereas CV for 2-hour glucose was lower for youth (P = .041). Classification concordance by HbA1c was lower in youth (P = .004). Using OGTT or HbA1c for classification, intervisit variability produced discordant classification in 20% of youth and 28% of adults. Using both fasting glucose and HbA1c, intervisit variability reduced discordant classification to 16% of adults while not improving classification in youth. CONCLUSIONS Poor reproducibility and lack of classification concordance highlight the limitations of one-time testing, with important implications for assessing eligibility in clinical trials. Consideration should be given to using more than a single parameter for screening and diagnosis, especially when classification category is important.
Collapse
Affiliation(s)
- Ashley H Tjaden
- The Biostatistics Center, Milken Institute School of Public Health The George Washington University, Rockville, MD, USA
| | - Sharon L Edelstein
- The Biostatistics Center, Milken Institute School of Public Health The George Washington University, Rockville, MD, USA
| | - Silva Arslanian
- Division of Pediatric Endocrinology, Diabetes, and Metabolism, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Elena Barengolts
- Department of Medicine, University of Illinois and Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Sonia Caprio
- Pediatric Endocrinology & Diabetes, Yale University School of Medicine, New Haven, CT, USA
| | - Melanie Cree-Green
- Pediatric Endocrinology, University of Colorado Anschutz Medical Campus/Children’s Hospital Colorado, Aurora, CO, USA
| | - Amale Lteif
- Division of Endocrinology and Metabolism, Indiana University School of Medicine and Roudebush VA Medical Center, Indianapolis, IN, USA
| | - Kieren J Mather
- Division of Endocrinology and Metabolism, Indiana University School of Medicine and Roudebush VA Medical Center, Indianapolis, IN, USA
| | - Mary Savoye
- Pediatric Endocrinology & Diabetes, Yale University School of Medicine, New Haven, CT, USA
| | - Anny H Xiang
- Department of Research & Evaluation, Kaiser Permanente Southern California, Los Angeles, CA, USA
| | - Steven E Kahn
- Division of Metabolism, Endocrinology and Nutrition, VA Puget Sound Health Care System and University of Washington, Seattle, WA, USA
| |
Collapse
|
29
|
TODAY Study Group, Nadeau KJ, El ghormli L, Arslanian S, Bacha F, Caprio S, Chan C, Chao LC, Rayas M, Siska MK, Zeitler P. Effect of Early Glycemic Control in Youth-Onset Type 2 Diabetes on Longer-Term Glycemic Control and β-Cell Function: Results From the TODAY Study. Diabetes Care 2023; 46:1507-1514. [PMID: 37378967 PMCID: PMC10369125 DOI: 10.2337/dc23-0560] [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: 03/29/2023] [Accepted: 05/15/2023] [Indexed: 06/29/2023]
Abstract
OBJECTIVE Little is known about the impact of early attainment of tight glycemic control on long-term β-cell function and glycemic control in youth-onset type 2 diabetes. We examined the effect of the initial 6 months of glycemic control on β-cell function and glycemic control longitudinally over 9 years and the impact of sex, race/ethnicity, and BMI on these relationships in adolescents with youth-onset type 2 diabetes in the Treatment Options for Type 2 Diabetes in Adolescents and Youth (TODAY) study. RESEARCH DESIGN AND METHODS Oral glucose tolerance tests were performed longitudinally through year 9 to derive estimates of insulin sensitivity and secretion. Early glycemia was defined by mean HbA1c during the first 6 months postrandomization, categorized into five HbA1c groups (<5.7%, 5.7 to <6.4%, 6.4 to <7.0%, 7.0 to <8.0%, and ≥8.0%). The long-term period was defined as the period between years 2 and 9. RESULTS A total of 656 participants (64.8% female, baseline mean age 14 years, diabetes duration <2 years) had longitudinal data available over an average of 6.4 ± 3.2 years of follow-up. HbA1c significantly increased in all early glycemic groups during years 2-9, with a steeper increase (+0.40%/year) among participants with the tightest initial control (mean early HbA1c <5.7%), in parallel to a decline in the C-peptide-derived disposition index. Nevertheless, the lower HbA1c categories continued to have relatively lower HbA1c over time. CONCLUSIONS Early tight glycemic control in the TODAY study was related to β-cell reserve and translated to better long-term glycemic control. However, tight early glycemic control on the randomized treatment in the TODAY study did not prevent deterioration of β-cell function.
Collapse
|
30
|
Kim G, Richards MC, Smith AB, Strybos KM, Huang X, Staggers K, Stayer SA, Baijal RG. Lack of Association of A1C With Postoperative Complications in Children With Type 1 or Type 2 Diabetes. Clin Diabetes 2023; 42:65-73. [PMID: 38230331 PMCID: PMC10788671 DOI: 10.2337/cd23-0002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
Not meeting recommended A1C targets may be associated with postoperative complications in adults, but there are no studies reporting on the relationship between preoperative A1C and postoperative complications in children with type 1 or type 2 diabetes. The objective of this study was to determine whether elevated A1C levels were associated with an increased incidence of postoperative complications in children with diabetes presenting for elective noncardiac surgery or diagnostic procedures. It found no such association, suggesting no need to delay elective surgery in children with diabetes until A1C is optimized.
Collapse
Affiliation(s)
- Grace Kim
- Department of Pediatrics, Division of Diabetes and Endocrinology, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX
| | | | | | | | - Xiaofan Huang
- Dan L. Duncan Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX
| | - Kristen Staggers
- Dan L. Duncan Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX
| | - Stephen A. Stayer
- Department of Pediatric Anesthesiology, Perioperative, and Pain Medicine, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX
| | - Rahul G. Baijal
- Department of Pediatric Anesthesiology, Perioperative, and Pain Medicine, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX
| |
Collapse
|
31
|
Wilson V. Managing type 2 diabetes in children and young people: challenges and solutions. Nurs Child Young People 2023; 35:35-42. [PMID: 37005859 DOI: 10.7748/ncyp.2023.e1460] [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] [Accepted: 08/19/2022] [Indexed: 04/04/2023]
Abstract
Growing numbers of children and young people are diagnosed with type 2 diabetes, partly due to the obesogenic environment they grow up in. The increasing prevalence of type 2 diabetes is seen particularly in adolescent girls and in children and young people of non-white ethnic backgrounds. There are numerous challenges relating to the diagnosis, treatment and management of type 2 diabetes in children and young people, notably the fact that the condition can lead to serious complications and often triggers high levels of anxiety and stress in patients and families. This article outlines the challenges faced by children and young people with type 2 diabetes, their families and carers, and suggests ways in which nurses can support them with the aim of achieving optimal management and self-management.
Collapse
|
32
|
Ha J, Chung ST, Springer M, Kim JY, Chen P, Cree MG, Behn CD, Sumner AE, Arslanian S, Sherman AS. Estimating Insulin Sensitivity and Beta-Cell Function from the Oral Glucose Tolerance Test: Validation of a new Insulin Sensitivity and Secretion (ISS) Model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.16.545377. [PMID: 37503271 PMCID: PMC10370185 DOI: 10.1101/2023.06.16.545377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Efficient and accurate methods to estimate insulin sensitivity (SI) and beta-cell function (BCF) are of great importance for studying the pathogenesis and treatment effectiveness of type 2 diabetes. Many methods exist, ranging in input data and technical requirements. Oral glucose tolerance tests (OGTTs) are preferred because they are simpler and more physiological. However, current analytical methods for OGTT-derived SI and BCF also range in complexity; the oral minimal models require mathematical expertise for deconvolution and fitting differential equations, and simple algebraic models (e.g., Matsuda index, insulinogenic index) may produce unphysiological values. We developed a new ISS (Insulin Secretion and Sensitivity) model for clinical research that provides precise and accurate estimates of SI and BCF from a standard OGTT, focusing on effectiveness, ease of implementation, and pragmatism. The model was developed by fitting a pair of differential equations to glucose and insulin without need of deconvolution or C-peptide data. The model is derived from a published model for longitudinal simulation of T2D progression that represents glucose-insulin homeostasis, including post-challenge suppression of hepatic glucose production and first- and second-phase insulin secretion. The ISS model was evaluated in three diverse cohorts including individuals at high risk of prediabetes (adult women with a wide range of BMI and adolescents with obesity). The new model had strong correlation with gold-standard estimates from intravenous glucose tolerance tests and hyperinsulinemic-euglycemic clamp. The ISS model has broad clinical applicability among diverse populations because it balances performance, fidelity, and complexity to provide a reliable phenotype of T2D risk.
Collapse
Affiliation(s)
- Joon Ha
- Department of Mathematics, Howard University, Washington, DC
| | - Stephanie T. Chung
- Section on Pediatric Diabetes, Obesity, and Metabolism, Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Max Springer
- Department of Mathematics, University of Maryland, College Park, MD
| | - Joon Young Kim
- Department of Exercise Science, David B. Falk College of Sport and Human Dynamics, Syracuse University, Syracuse, NY
| | | | - Melanie G. Cree
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Cecilia Diniz Behn
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO
- Department of Applied Mathematics and Statistics, Colorado School of Mines, Golden, Colorado
| | - Anne E. Sumner
- Intramural Research Program, National Institute on Minority Health and Health Disparities (NIMHD), National Institutes of Health, Bethesda, MD
- Section on Ethnicity and Health, Diabetes Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD
| | - Silva Arslanian
- Division of Pediatric Endocrinology, Metabolism and Diabetes Mellitus, Center for Pediatric Research in Obesity and Metabolism, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA
| | - Arthur S. Sherman
- Laboratory of Biological Modeling, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, MD
| |
Collapse
|
33
|
Fan Y, S H Lau E, Wu H, Yang A, Chow E, P S Kong A, C W Ma R, C N Chan J, O Y Luk A. Higher incidence of cardiovascular-kidney complications in Chinese with youth-onset type 2 diabetes versus youth-onset type 1 diabetes attenuated by control of cardio-metabolic risk factors: a population-based prospective cohort study in Hong Kong. Diabetes Res Clin Pract 2023:110728. [PMID: 37217017 DOI: 10.1016/j.diabres.2023.110728] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 05/09/2023] [Accepted: 05/18/2023] [Indexed: 05/24/2023]
Abstract
AIMS To determine and compare the incidence of diabetes complications in Chinese with youth-onset type 2 and type 1 diabetes. METHODS We conducted a population-based prospective cohort study, including 1,260 people with type 2 diabetes and 1,227 with type 1 diabetes diagnosed at age <20 years who underwent metabolic and complication assessment in Hong Kong Hospital Authority between 2000 and 2018. They were followed for incident cardiovascular disease (CVD), end-stage kidney disease (ESKD) and all-cause death until 2019. Multivariable Cox regression analysis was applied to compare the risks of these complications in type 2 versus type 1 diabetes. RESULTS People with type 1 diabetes (median age: 20 years, median diabetes duration: 9 years) and type 2 diabetes (median age: 21 years, median diabetes duration: 6 years) were followed for a mean period of 9.2 and 8.8 years respectively. The risks of CVD (HR [95% CI] 1.66 [1.01-2.72]) and ESKD (HR 1.96 [1.27-3.04]) but not death (HR 1.10 [0.72-1.67]) were higher in type 2 versus type 1 diabetes, adjusted for age at diagnosis, diabetes duration and sex. The association became nonsignificant with further adjustment for glycaemic and metabolic control. Youth-onset type 2 diabetes conferred mortality excess (standardized mortality ratio 4.15 [3.28-5.17]) to age- and sex-matched general population. CONCLUSIONS People with youth-onset type 2 diabetes had higher incidence rates of CVD and ESKD than type 1 diabetes. The excess risks in type 2 diabetes were removed after adjusted for cardio-metabolic risk factors.
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
| | - 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
| | - Hongjiang Wu
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Aimin Yang
- 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
| | - 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; Hong Kong Institute of Diabetes and Obesity, 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
| | - 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; Hong Kong Institute of Diabetes and Obesity, 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
| | - 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; Hong Kong Institute of Diabetes and Obesity, 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
| | - 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; Hong Kong Institute of Diabetes and Obesity, 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.
| |
Collapse
|
34
|
Serbis A, Giapros V, Tsamis K, Balomenou F, Galli-Tsinopoulou A, Siomou E. Beta Cell Dysfunction in Youth- and Adult-Onset Type 2 Diabetes: An Extensive Narrative Review with a Special Focus on the Role of Nutrients. Nutrients 2023; 15:2217. [PMID: 37432389 PMCID: PMC10180650 DOI: 10.3390/nu15092217] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 05/04/2023] [Accepted: 05/06/2023] [Indexed: 07/12/2023] Open
Abstract
Traditionally a disease of adults, type 2 diabetes (T2D) has been increasingly diagnosed in youth, particularly among adolescents and young adults of minority ethnic groups. Especially, during the recent COVID-19 pandemic, obesity and prediabetes have surged not only in minority ethnic groups but also in the general population, further raising T2D risk. Regarding its pathogenesis, a gradually increasing insulin resistance due to central adiposity combined with a progressively defective β-cell function are the main culprits. Especially in youth-onset T2D, a rapid β-cell activity decline has been observed, leading to higher treatment failure rates, and early complications. In addition, it is well established that both the quantity and quality of food ingested by individuals play a key role in T2D pathogenesis. A chronic imbalance between caloric intake and expenditure together with impaired micronutrient intake can lead to obesity and insulin resistance on one hand, and β-cell failure and defective insulin production on the other. This review summarizes our evolving understanding of the pathophysiological mechanisms involved in defective insulin secretion by the pancreatic islets in youth- and adult-onset T2D and, further, of the role various micronutrients play in these pathomechanisms. This knowledge is essential if we are to curtail the serious long-term complications of T2D both in pediatric and adult populations.
Collapse
Affiliation(s)
- Anastasios Serbis
- Department of Pediatrics, School of Medicine, University of Ioannina, St. Niarhcos Avenue, 45500 Ioannina, Greece;
| | - Vasileios Giapros
- Neonatal Intensive Care Unit, School of Medicine, University of Ioannina, St. Νiarhcos Avenue, 45500 Ioannina, Greece (F.B.)
| | - Konstantinos Tsamis
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, St. Niarhcos Avenue, 45500 Ioannina, Greece
| | - Foteini Balomenou
- Neonatal Intensive Care Unit, School of Medicine, University of Ioannina, St. Νiarhcos Avenue, 45500 Ioannina, Greece (F.B.)
| | - Assimina Galli-Tsinopoulou
- Second Department of Pediatrics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, AHEPA University Hospital, Stilponos Kyriakidi 1, 54636 Thessaloniki, Greece;
| | - Ekaterini Siomou
- Department of Pediatrics, School of Medicine, University of Ioannina, St. Niarhcos Avenue, 45500 Ioannina, Greece;
| |
Collapse
|
35
|
Rodriquez IM, O’Sullivan KL. Youth-Onset Type 2 Diabetes: Burden of Complications and Socioeconomic Cost. Curr Diab Rep 2023; 23:59-67. [PMID: 36961664 PMCID: PMC10037371 DOI: 10.1007/s11892-023-01501-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/08/2023] [Indexed: 03/25/2023]
Abstract
PURPOSE OF REVIEW With the rise in prevalence of youth-onset type 2 diabetes (T2DM), it is imperative to understand the clinical burden of the disease and the socioeconomic burden this disease imposes. We review the most recent data on youth-onset T2DM, including its pathophysiology, complications, and treatment. We also review existing data to determine the socioeconomic burden of youth-onset T2DM. RECENT FINDINGS The incidence of youth-onset T2DM is rising, and significantly accelerated following the COVID-19 pandemic. Youth with T2DM are more frequently from families of racial/ethnic minorities and lower socioeconomic status. Youth-onset T2DM has more rapid disease progression compared to adult-onset type 2 diabetes. It results in earlier and more severe microvascular and macrovascular complications compared to both adult-onset T2DM and youth-onset type 1 diabetes (T1DM). While there is a lack of data describing the socioeconomic cost of youth-onset T2DM, based on extrapolation from analyses of the burden of T2DM in adults and T1DM in youth, we propose that youth-onset T2DM has higher direct and indirect costs than adult-onset T2DM. Youth-onset T2DM presents a significant clinical and socioeconomic burden due to its aggressive presentation and earlier appearance of complications. Additional research is needed regarding the cost of illness in this population.
Collapse
Affiliation(s)
- Isabella Marranzini Rodriquez
- Medical Center, Section of Adult & Pediatric Endocrinology, Diabetes & Metabolism, University of Chicago, 5841 S. Maryland Avenue, MC 5053, Chicago, IL 60637 USA
| | - Katie L. O’Sullivan
- Medical Center, Section of Adult & Pediatric Endocrinology, Diabetes & Metabolism, University of Chicago, 5841 S. Maryland Avenue, MC 5053, Chicago, IL 60637 USA
| |
Collapse
|
36
|
Bjornstad P, Chao LC, Cree-Green M, Dart AB, King M, Looker HC, Magliano DJ, Nadeau KJ, Pinhas-Hamiel O, Shah AS, van Raalte DH, Pavkov ME, Nelson RG. Youth-onset type 2 diabetes mellitus: an urgent challenge. Nat Rev Nephrol 2023; 19:168-184. [PMID: 36316388 PMCID: PMC10182876 DOI: 10.1038/s41581-022-00645-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/03/2022] [Indexed: 11/05/2022]
Abstract
The incidence and prevalence of youth-onset type 2 diabetes mellitus (T2DM) and its complications are increasing worldwide. Youth-onset T2DM has been reported in all racial and ethnic groups, but Indigenous peoples and people of colour are disproportionately affected. People with youth-onset T2DM often have a more aggressive clinical course than those with adult-onset T2DM or those with type 1 diabetes mellitus. Moreover, the available treatment options for children and adolescents with T2DM are more limited than for adult patients. Intermediate complications of youth-onset T2DM, such as increased albuminuria, often develop in late childhood or early adulthood, and end-stage complications, including kidney failure, develop in mid-life. The increasing frequency, earlier onset and greater severity of childhood obesity in the past 50 years together with increasingly sedentary lifestyles and an increasing frequency of intrauterine exposure to diabetes are important drivers of the epidemic of youth-onset T2DM. The particularly high risk of the disease in historically disadvantaged populations suggests an important contribution of social and environmental factors, including limited access to high-quality health care, healthy food choices and opportunities for physical activity as well as exposure to stressors including systemic racism and environmental pollutants. Understanding the mechanisms that underlie the development and aggressive clinical course of youth-onset T2DM is key to identifying successful prevention and management strategies.
Collapse
Affiliation(s)
| | - Lily C Chao
- Children's Hospital Los Angeles, University of Southern California, Keck School of Medicine, Los Angeles, CA, USA
| | | | - Allison B Dart
- Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Canada
| | - Malcolm King
- University of Saskatchewan College of Medicine, Saskatoon, Saskatchewan, Canada
| | - Helen C Looker
- National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ, USA
| | - Dianna J Magliano
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Monash University, School of Public Health and Preventive Medicine, Melbourne, Australia
| | | | - Orit Pinhas-Hamiel
- Paediatric Endocrine and Diabetes Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Amy S Shah
- Cincinnati Children's Hospital and The University of Cincinnati, Cincinnati, OH, USA
| | | | - Meda E Pavkov
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Robert G Nelson
- National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ, USA.
| |
Collapse
|
37
|
Hao S, Umpierrez GE, Daley T, Vellanki P. Intervention with Therapeutic Agents, Understanding the Path to Remission in Type 2 Diabetes: Part 1. Endocrinol Metab Clin North Am 2023; 52:27-38. [PMID: 36754495 DOI: 10.1016/j.ecl.2022.07.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Type 2 diabetes is characterized by progressive decline in pancreatic β-cell function. Studies in adult subjects with newly diagnosed type 2 diabetes have reported that intensive insulin therapy followed by various antihyperglycemic medications can delay β-cell decline. However, this improvement is lost after cessation of therapy. In contrast, youth with type 2 diabetes experience a more rapid loss in β-cell function compared with adults and have loss of β-cell function despite being on insulin and other antihyperglycemic medications. In part one of this two-part review, we discuss studies aiming to achieve diabetes remission with insulin and oral antidiabetic medications.
Collapse
Affiliation(s)
- Shuai Hao
- Division of Pediatric Endocrinology, Children's Healthcare of Atlanta, Emory University School of Medicine, 1400 Tullie Road Northeast, Atlanta, GA 30329, USA
| | - Guillermo E Umpierrez
- Division of Endocrinology, Metabolism & Lipids, Emory University School of Medicine, 69 Jesse Hill Jr Drive Southeast, Glenn Building, Room 205, Suite 200, Atlanta, GA 30303, USA
| | - Tanicia Daley
- Division of Pediatric Endocrinology, Children's Healthcare of Atlanta, Emory University School of Medicine, 1400 Tullie Road Northeast, Atlanta, GA 30329, USA
| | - Priyathama Vellanki
- Division of Endocrinology, Metabolism & Lipids, Emory University School of Medicine, 69 Jesse Hill Jr Drive Southeast, Glenn Building, Room 205, Suite 200, Atlanta, GA 30303, USA.
| |
Collapse
|
38
|
Zhang X, Wu H, Fan B, Shi M, Lau ES, Yang A, Chow E, Kong AP, Chan JC, Ma RC, Luk AO. The role of age on the risk relationship between prediabetes and major morbidities and mortality: Analysis of the Hong Kong diabetes surveillance database of 2 million Chinese adults. THE LANCET REGIONAL HEALTH. WESTERN PACIFIC 2023; 30:100599. [PMID: 36419741 PMCID: PMC9677132 DOI: 10.1016/j.lanwpc.2022.100599] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
BACKGROUND Intensive lifestyle modification showed variable success in the prevention of major clinical events and mortality among people with prediabetes. We propose that age may partly explain the heterogeneity and that health hazards related to prediabetes are age-specific. METHODS We conducted a retrospective analysis of a territory-wide diabetes surveillance dataset from the Hong Kong Hospital Authority between 2000 and 2019. Prediabetes was defined according to the American Diabetes Association criteria. Proportional Cox regression was performed, stratified by baseline age categories (20-39, 40-59, 60-79 and ≥80 years). FINDINGS 1,630,942 individuals were included in the analysis. Compared with normoglycaemia, prediabetes was associated with greater hazards for cardiovascular disease (CVD) and all-cause mortality in most age groups but the effect size attenuated with ascending age (p value for trend <0·05). In the youngest and in the oldest age categories, the respective hazard ratios (95% confidence interval) of prediabetes vs normoglycaemia were 1·79 (1·59, 2·01) and 1·00 (0·95, 1·05) for CVD, and 1·36 (1·20, 1·55) and 0·99 (0·97, 1·02) for all-cause mortality. Similar associations were found for chronic kidney disease, end-stage kidney disease, all-site cancer, all-site infection, subtypes of CVD, and cause-specific mortality. The associations became attenuated but remained after excluding people who later developed diabetes and adjusting for metabolic factors. Similar associations were observed in prediabetes defined by impaired fasting glucose, but not HbA1c. INTERPRETATION Prediabetes is associated with higher risk of major clinical events, even excluding subsequent development of diabetes and adjusting for metabolic factors. The risk relationships are stronger in young than older people. FUNDING This study did not receive any specific funding.
Collapse
Affiliation(s)
- Xinge Zhang
- Department of Medicine and Therapeutics, 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
| | - Baoqi Fan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Mai Shi
- 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
| | - Aimin Yang
- 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
| | - 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
- Hong Kong Institute of Diabetes and Obesity, 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
| | - 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
- Hong Kong Institute of Diabetes and Obesity, 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
| | - 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
- Hong Kong Institute of Diabetes and Obesity, 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
| | - 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
- Hong Kong Institute of Diabetes and Obesity, 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
| |
Collapse
|
39
|
Shah AS, Zeitler PS, Wong J, Pena AS, Wicklow B, Arslanian S, Chang N, Fu J, Dabadghao P, Pinhas-Hamiel O, Urakami T, Craig ME. ISPAD Clinical Practice Consensus Guidelines 2022: Type 2 diabetes in children and adolescents. Pediatr Diabetes 2022; 23:872-902. [PMID: 36161685 DOI: 10.1111/pedi.13409] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 08/29/2022] [Indexed: 11/29/2022] Open
Abstract
Since the 2018 ISPAD guidelines on this topic, follow-up of large cohorts from around the globe have continued informing the current incidence and prevalence of co-morbidities and complications in young adults with youth-onset type 2 diabetes (T2D). This chapter focuses on the risk factors, diagnosis and presentation of youth-onset T2D, the initial and subsequent management of youth-onset T2D, and management of co-morbidities and complications. We include key updates from the observational phase of the multi-center Treatment Options for Type 2 Diabetes in Adolescents and Youth (TODAY) clinical trial, the SEARCH for Diabetes in Youth (SEARCH) study and new data from the Restoring Insulin Secretion (RISE) study, a head-to-head comparison of youth onset vs adult-onset T2D. We also include an expanded section on risk factors associated with T2D, algorithms and tables for treatment, management, and assessment of co-morbidities and complications, and sections on recently approved pharmacologic therapies for the treatment of youth-onset T2D, social determinants of health, and settings of care given COVID-19 pandemic.
Collapse
Affiliation(s)
- Amy S Shah
- Division of Pediatric Endocrinology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati, Cincinnati, Ohio, USA
| | - Philip S Zeitler
- Division of Pediatric Endocrinology, Children's Hospital Colorado and University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Jencia Wong
- Department of Endocrinology, Royal Prince Alfred Hospital and Central Clinical School, Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Alexia S Pena
- The University of Adelaide, Robinson Research Institute, North Adelaide, South Australia, Australia
| | - Brandy Wicklow
- Division of Endocrinology, Winnipeg Children's Hospital and University of Manitoba, Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Silva Arslanian
- Division of Pediatric Endocrinology, Metabolism, and Diabetes Mellitus, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Nancy Chang
- Center for Endocrinology, Diabetes and Metabolism, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Junfen Fu
- Division of Endocrinology, The Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Preeti Dabadghao
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Orit Pinhas-Hamiel
- Edmond and Lily Safra Children's Hospital, Sackler School of Medicine, Tel-Aviv, Israel
| | - Tatsuhiko Urakami
- Department of Pediatrics, Nihon University School of Medicine, Tokyo, Japan
| | - Maria E Craig
- The Children's Hospital at Westmead, University of Sydney, Sydney, New South Wales, Australia
- Discipline of Pediatrics & Child Health, School of Clinical Medicine, University of NSW Medicine and Health, Sydney, New South Wales, Australia
| |
Collapse
|
40
|
Isganaitis E, Laffel L. Recommendations for Screening Children and Adolescents for Prediabetes and Type 2 Diabetes. JAMA 2022; 328:933-934. [PMID: 36098736 DOI: 10.1001/jama.2022.13759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Elvira Isganaitis
- Pediatric, Adolescent and Young Adult Section, Harvard Medical School, Boston, Massachusetts
- Section on Clinical, Behavioral and Outcomes Research, Harvard Medical School
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - Lori Laffel
- Pediatric, Adolescent and Young Adult Section, Harvard Medical School, Boston, Massachusetts
- Section on Clinical, Behavioral and Outcomes Research, Harvard Medical School
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
41
|
Fan Y, Lau ESH, Wu H, Yang A, Chow E, So WY, Kong APS, Ma RCW, Chan JCN, Luk AOY. Incidence of long-term diabetes complications and mortality in youth-onset type 2 diabetes: A systematic review. Diabetes Res Clin Pract 2022; 191:110030. [PMID: 35934175 DOI: 10.1016/j.diabres.2022.110030] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 06/17/2022] [Accepted: 08/01/2022] [Indexed: 11/16/2022]
Abstract
AIMS This systematic review aims to assess the incidence of chronic kidney disease (CKD), cardiovascular disease (CVD) and mortality in people with type 2 diabetes diagnosed <20 years. METHODS We searched MEDLINE, Embase and Cochrane Library for longitudinal studies published between 1 January 2000 and 31 November 2021. RESULTS Seventeen studies (15 reporting CKD, 3 reporting CVD, 5 reporting mortality) from seven countries of sample size ranging between 96 and 4,141 were eligible. Most studies were conducted in North America and Europe (n = 14). Diabetes duration at enrolment varied from 0 to 8.3 years and follow-up duration from 1 to 12.6 years. The incidence rates (per 1,000 person-year) of albuminuria ranged between 12.4 and 114.8, macroalbuminuria or proteinuria between 10 and 35.0, end-stage kidney disease (ESKD) between 0.4 and 25.0, CVD between 3.7 and 19.5, and mortality between 1.0 and 18.6. The highest incidence rates of albuminuria, ESKD and mortality were recorded in Australian Aboriginal and Pima Indian populations. Youth-onset type 2 diabetes was associated with greater risk of developing CKD compared with type 1 diabetes in most studies. CONCLUSION Studies reporting CVD in youth-onset type 2 diabetes are scarce. Estimated incidence rates of CKD and mortality in youth-onset type 2 diabetes varied across different study populations, potentially higher in indigenous people. Youth with type 2 diabetes are at higher risk of adverse kidney outcomes than their type 1 counterparts. More studies are needed in regions outside of North America and Europe.
Collapse
Affiliation(s)
- Yingnan Fan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Eric S H Lau
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Hongjiang Wu
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Aimin Yang
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Elaine Chow
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Wing-Yee So
- Hong Kong Hospital Authority, Kowloon, Hong Kong, China
| | - Alice P S Kong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong, China; Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Shatin, Hong Kong, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Ronald C W Ma
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong, China; Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Shatin, Hong Kong, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Juliana C N Chan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong, China; Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Shatin, Hong Kong, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Andrea O Y Luk
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong, China; Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Shatin, Hong Kong, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| |
Collapse
|
42
|
Hampton GS, Bartlette K, Nadeau KJ, Cree-Green M, Diniz Behn C. Mathematical modeling reveals differential dynamics of insulin action models on glycerol and glucose in adolescent girls with obesity. Front Physiol 2022; 13:895118. [PMID: 35991189 PMCID: PMC9388790 DOI: 10.3389/fphys.2022.895118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 07/08/2022] [Indexed: 12/30/2022] Open
Abstract
Under healthy conditions, the pancreas responds to a glucose challenge by releasing insulin. Insulin suppresses lipolysis in adipose tissue, thereby decreasing plasma glycerol concentration, and it regulates plasma glucose concentration through action in muscle and liver. Insulin resistance (IR) occurs when more insulin is required to achieve the same effects, and IR may be tissue-specific. IR emerges during puberty as a result of high concentrations of growth hormone and is worsened by youth-onset obesity. Adipose, liver, and muscle tissue exhibit distinct dose-dependent responses to insulin in multi-phase hyperinsulinemic-euglycemic (HE) clamps, but the HE clamp protocol does not address potential differences in the dynamics of tissue-specific insulin responses. Changes to the dynamics of insulin responses would alter glycemic control in response to a glucose challenge. To investigate the dynamics of insulin acting on adipose tissue, we developed a novel differential-equations based model that describes the coupled dynamics of glycerol concentrations and insulin action during an oral glucose tolerance test in female adolescents with obesity and IR. We compared these dynamics to the dynamics of insulin acting on muscle and liver as assessed with the oral minimal model applied to glucose and insulin data collected under the same protocol. We found that the action of insulin on glycerol peaks approximately 67 min earlier (p < 0.001) and follows the dynamics of plasma insulin more closely compared to insulin action on glucose as assessed by the parameters representing the time constants for insulin action on glucose and glycerol (p < 0.001). These findings suggest that the dynamics of insulin action show tissue-specific differences in our IR adolescent population, with adipose tissue responding to insulin more quickly compared to muscle and liver. Improved understanding of the tissue-specific dynamics of insulin action may provide novel insights into the progression of metabolic disease in patient populations with diverse metabolic phenotypes.
Collapse
Affiliation(s)
- Griffin S. Hampton
- Department of Applied Mathematics and Statistics, Colorado School of Mines, Golden, CO, United States
| | - Kai Bartlette
- Department of Applied Mathematics and Statistics, Colorado School of Mines, Golden, CO, United States
| | - Kristen J. Nadeau
- Division of Pediatric Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States,Ludeman Center for Women’s Health Research, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Melanie Cree-Green
- Division of Pediatric Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States,Ludeman Center for Women’s Health Research, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Cecilia Diniz Behn
- Department of Applied Mathematics and Statistics, Colorado School of Mines, Golden, CO, United States,Division of Pediatric Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States,*Correspondence: Cecilia Diniz Behn,
| |
Collapse
|
43
|
Arslanian SA, Hannon T, Zeitler P, Chao LC, Boucher-Berry C, Barrientos-Pérez M, Bismuth E, Dib S, Cho JI, Cox D. Once-Weekly Dulaglutide for the Treatment of Youths with Type 2 Diabetes. N Engl J Med 2022; 387:433-443. [PMID: 35658022 DOI: 10.1056/nejmoa2204601] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND The incidence of type 2 diabetes mellitus is increasing among youths. Once-weekly treatment with dulaglutide, a glucagon-like peptide-1 receptor agonist, may have efficacy with regard to glycemic control in youths with type 2 diabetes. METHODS In a double-blind, placebo-controlled, 26-week trial, we randomly assigned participants (10 to <18 years of age; body-mass index [BMI], >85th percentile) being treated with lifestyle modifications alone or with metformin, with or without basal insulin, in a 1:1:1 ratio to receive once-weekly subcutaneous injections of placebo, dulaglutide at a dose of 0.75 mg, or dulaglutide at a dose of 1.5 mg. Participants were then included in a 26-week open-label extension study in which those who had received placebo began receiving dulaglutide at a weekly dose of 0.75 mg. The primary end point was the change from baseline in the glycated hemoglobin level at 26 weeks. Secondary end points included a glycated hemoglobin level of less than 7.0% and changes from baseline in the fasting glucose concentration and BMI. Safety was also assessed. RESULTS A total of 154 participants underwent randomization. At 26 weeks, the mean glycated hemoglobin level had increased in the placebo group (0.6 percentage points) and had decreased in the dulaglutide groups (-0.6 percentage points in the 0.75-mg group and -0.9 percentage points in the 1.5-mg group, P<0.001 for both comparisons vs. placebo). At 26 weeks, a higher percentage of participants in the pooled dulaglutide groups than in the placebo group had a glycated hemoglobin level of less than 7.0% (51% vs. 14%, P<0.001). The fasting glucose concentration increased in the placebo group (17.1 mg per deciliter) and decreased in the pooled dulaglutide groups (-18.9 mg per deciliter, P<0.001), and there were no between-group differences in the change in BMI. The incidence of gastrointestinal adverse events was higher with dulaglutide therapy than with placebo. The safety profile of dulaglutide was consistent with that reported in adults. CONCLUSIONS Treatment with dulaglutide at a once-weekly dose of 0.75 mg or 1.5 mg was superior to placebo in improving glycemic control through 26 weeks among youths with type 2 diabetes who were being treated with or without metformin or basal insulin, without an effect on BMI. (Funded by Eli Lilly; AWARD-PEDS ClinicalTrials.gov number, NCT02963766.).
Collapse
Affiliation(s)
- Silva A Arslanian
- From the Center for Pediatric Research in Obesity and Metabolism, Division of Pediatric Endocrinology, Diabetes, and Metabolism, University of Pittsburgh, School of Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh (S.A.A.); the Department of Pediatrics, Indiana University School of Medicine, Riley Hospital for Children (T.H.), and Eli Lilly (J.I.C., D.C.) - both in Indianapolis; Children's Hospital Colorado, University of Colorado School of Medicine, Aurora (P.Z.); Children's Hospital Los Angeles, Los Angeles (L.C.C.); the Department of Pediatric Endocrinology, University of Illinois Medical Center at Chicago, Chicago (C.B.-B.); the Division of Pediatric Endocrinology, Hospital Ángeles Puebla, Puebla City, Mexico (M.B.-P.); the Department of Pediatric Endocrinology, Assistance Publique-Hôpitaux de Paris, Robert Debré Hospital, Paris (E.B.); and the Diabetes Center of Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo (S.D.)
| | - Tamara Hannon
- From the Center for Pediatric Research in Obesity and Metabolism, Division of Pediatric Endocrinology, Diabetes, and Metabolism, University of Pittsburgh, School of Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh (S.A.A.); the Department of Pediatrics, Indiana University School of Medicine, Riley Hospital for Children (T.H.), and Eli Lilly (J.I.C., D.C.) - both in Indianapolis; Children's Hospital Colorado, University of Colorado School of Medicine, Aurora (P.Z.); Children's Hospital Los Angeles, Los Angeles (L.C.C.); the Department of Pediatric Endocrinology, University of Illinois Medical Center at Chicago, Chicago (C.B.-B.); the Division of Pediatric Endocrinology, Hospital Ángeles Puebla, Puebla City, Mexico (M.B.-P.); the Department of Pediatric Endocrinology, Assistance Publique-Hôpitaux de Paris, Robert Debré Hospital, Paris (E.B.); and the Diabetes Center of Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo (S.D.)
| | - Philip Zeitler
- From the Center for Pediatric Research in Obesity and Metabolism, Division of Pediatric Endocrinology, Diabetes, and Metabolism, University of Pittsburgh, School of Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh (S.A.A.); the Department of Pediatrics, Indiana University School of Medicine, Riley Hospital for Children (T.H.), and Eli Lilly (J.I.C., D.C.) - both in Indianapolis; Children's Hospital Colorado, University of Colorado School of Medicine, Aurora (P.Z.); Children's Hospital Los Angeles, Los Angeles (L.C.C.); the Department of Pediatric Endocrinology, University of Illinois Medical Center at Chicago, Chicago (C.B.-B.); the Division of Pediatric Endocrinology, Hospital Ángeles Puebla, Puebla City, Mexico (M.B.-P.); the Department of Pediatric Endocrinology, Assistance Publique-Hôpitaux de Paris, Robert Debré Hospital, Paris (E.B.); and the Diabetes Center of Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo (S.D.)
| | - Lily C Chao
- From the Center for Pediatric Research in Obesity and Metabolism, Division of Pediatric Endocrinology, Diabetes, and Metabolism, University of Pittsburgh, School of Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh (S.A.A.); the Department of Pediatrics, Indiana University School of Medicine, Riley Hospital for Children (T.H.), and Eli Lilly (J.I.C., D.C.) - both in Indianapolis; Children's Hospital Colorado, University of Colorado School of Medicine, Aurora (P.Z.); Children's Hospital Los Angeles, Los Angeles (L.C.C.); the Department of Pediatric Endocrinology, University of Illinois Medical Center at Chicago, Chicago (C.B.-B.); the Division of Pediatric Endocrinology, Hospital Ángeles Puebla, Puebla City, Mexico (M.B.-P.); the Department of Pediatric Endocrinology, Assistance Publique-Hôpitaux de Paris, Robert Debré Hospital, Paris (E.B.); and the Diabetes Center of Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo (S.D.)
| | - Claudia Boucher-Berry
- From the Center for Pediatric Research in Obesity and Metabolism, Division of Pediatric Endocrinology, Diabetes, and Metabolism, University of Pittsburgh, School of Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh (S.A.A.); the Department of Pediatrics, Indiana University School of Medicine, Riley Hospital for Children (T.H.), and Eli Lilly (J.I.C., D.C.) - both in Indianapolis; Children's Hospital Colorado, University of Colorado School of Medicine, Aurora (P.Z.); Children's Hospital Los Angeles, Los Angeles (L.C.C.); the Department of Pediatric Endocrinology, University of Illinois Medical Center at Chicago, Chicago (C.B.-B.); the Division of Pediatric Endocrinology, Hospital Ángeles Puebla, Puebla City, Mexico (M.B.-P.); the Department of Pediatric Endocrinology, Assistance Publique-Hôpitaux de Paris, Robert Debré Hospital, Paris (E.B.); and the Diabetes Center of Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo (S.D.)
| | - Margarita Barrientos-Pérez
- From the Center for Pediatric Research in Obesity and Metabolism, Division of Pediatric Endocrinology, Diabetes, and Metabolism, University of Pittsburgh, School of Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh (S.A.A.); the Department of Pediatrics, Indiana University School of Medicine, Riley Hospital for Children (T.H.), and Eli Lilly (J.I.C., D.C.) - both in Indianapolis; Children's Hospital Colorado, University of Colorado School of Medicine, Aurora (P.Z.); Children's Hospital Los Angeles, Los Angeles (L.C.C.); the Department of Pediatric Endocrinology, University of Illinois Medical Center at Chicago, Chicago (C.B.-B.); the Division of Pediatric Endocrinology, Hospital Ángeles Puebla, Puebla City, Mexico (M.B.-P.); the Department of Pediatric Endocrinology, Assistance Publique-Hôpitaux de Paris, Robert Debré Hospital, Paris (E.B.); and the Diabetes Center of Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo (S.D.)
| | - Elise Bismuth
- From the Center for Pediatric Research in Obesity and Metabolism, Division of Pediatric Endocrinology, Diabetes, and Metabolism, University of Pittsburgh, School of Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh (S.A.A.); the Department of Pediatrics, Indiana University School of Medicine, Riley Hospital for Children (T.H.), and Eli Lilly (J.I.C., D.C.) - both in Indianapolis; Children's Hospital Colorado, University of Colorado School of Medicine, Aurora (P.Z.); Children's Hospital Los Angeles, Los Angeles (L.C.C.); the Department of Pediatric Endocrinology, University of Illinois Medical Center at Chicago, Chicago (C.B.-B.); the Division of Pediatric Endocrinology, Hospital Ángeles Puebla, Puebla City, Mexico (M.B.-P.); the Department of Pediatric Endocrinology, Assistance Publique-Hôpitaux de Paris, Robert Debré Hospital, Paris (E.B.); and the Diabetes Center of Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo (S.D.)
| | - Sergio Dib
- From the Center for Pediatric Research in Obesity and Metabolism, Division of Pediatric Endocrinology, Diabetes, and Metabolism, University of Pittsburgh, School of Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh (S.A.A.); the Department of Pediatrics, Indiana University School of Medicine, Riley Hospital for Children (T.H.), and Eli Lilly (J.I.C., D.C.) - both in Indianapolis; Children's Hospital Colorado, University of Colorado School of Medicine, Aurora (P.Z.); Children's Hospital Los Angeles, Los Angeles (L.C.C.); the Department of Pediatric Endocrinology, University of Illinois Medical Center at Chicago, Chicago (C.B.-B.); the Division of Pediatric Endocrinology, Hospital Ángeles Puebla, Puebla City, Mexico (M.B.-P.); the Department of Pediatric Endocrinology, Assistance Publique-Hôpitaux de Paris, Robert Debré Hospital, Paris (E.B.); and the Diabetes Center of Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo (S.D.)
| | - Jang Ik Cho
- From the Center for Pediatric Research in Obesity and Metabolism, Division of Pediatric Endocrinology, Diabetes, and Metabolism, University of Pittsburgh, School of Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh (S.A.A.); the Department of Pediatrics, Indiana University School of Medicine, Riley Hospital for Children (T.H.), and Eli Lilly (J.I.C., D.C.) - both in Indianapolis; Children's Hospital Colorado, University of Colorado School of Medicine, Aurora (P.Z.); Children's Hospital Los Angeles, Los Angeles (L.C.C.); the Department of Pediatric Endocrinology, University of Illinois Medical Center at Chicago, Chicago (C.B.-B.); the Division of Pediatric Endocrinology, Hospital Ángeles Puebla, Puebla City, Mexico (M.B.-P.); the Department of Pediatric Endocrinology, Assistance Publique-Hôpitaux de Paris, Robert Debré Hospital, Paris (E.B.); and the Diabetes Center of Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo (S.D.)
| | - David Cox
- From the Center for Pediatric Research in Obesity and Metabolism, Division of Pediatric Endocrinology, Diabetes, and Metabolism, University of Pittsburgh, School of Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh (S.A.A.); the Department of Pediatrics, Indiana University School of Medicine, Riley Hospital for Children (T.H.), and Eli Lilly (J.I.C., D.C.) - both in Indianapolis; Children's Hospital Colorado, University of Colorado School of Medicine, Aurora (P.Z.); Children's Hospital Los Angeles, Los Angeles (L.C.C.); the Department of Pediatric Endocrinology, University of Illinois Medical Center at Chicago, Chicago (C.B.-B.); the Division of Pediatric Endocrinology, Hospital Ángeles Puebla, Puebla City, Mexico (M.B.-P.); the Department of Pediatric Endocrinology, Assistance Publique-Hôpitaux de Paris, Robert Debré Hospital, Paris (E.B.); and the Diabetes Center of Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo (S.D.)
| |
Collapse
|
44
|
Utzschneider KM, Ehrmann DA, Arslanian SA, Barengolts E, Buchanan TA, Caprio S, Edelstein SL, Hannon TS, Kahn SE, Kozedub A, Mather KJ, Nadeau KJ, Sam S, Tripputi M, Xiang AH, El ghormli L, The RISE Consortium. Weight loss and β-cell responses following gastric banding or pharmacotherapy in adults with impaired glucose tolerance or type 2 diabetes: a randomized trial. Obesity (Silver Spring) 2022; 30:1579-1588. [PMID: 35894078 PMCID: PMC10849844 DOI: 10.1002/oby.23475] [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: 02/10/2022] [Revised: 03/22/2022] [Accepted: 04/19/2022] [Indexed: 12/30/2022]
Abstract
OBJECTIVE The extent to which weight loss contributes to increases in insulin sensitivity (IS) and β-cell function after surgical or medical intervention has not been directly compared in individuals with impaired glucose tolerance or newly diagnosed type 2 diabetes. METHODS The Restoring Insulin Secretion (RISE) Study included adults in the Beta-Cell Restoration Through Fat Mitigation Study (n = 88 randomized to laparoscopic gastric banding or metformin [MET]) and the Adult Medication Study (n = 267 randomized to placebo, MET, insulin glargine/MET, or liraglutide + MET [L + M]). IS and β-cell responses were measured at baseline and after 12 months by modeling of oral glucose tolerance tests and during arginine-stimulated hyperglycemic clamps. Linear regression models assessed differences between and within treatments over time. RESULTS BMI decreased in all treatment groups, except placebo, at 12 months. IS increased in all arms except placebo and was inversely correlated with changes in BMI. L + M was the only treatment arm that enhanced multiple measures of β-cell function independent of weight loss. Insulin secretion decreased in the laparoscopic gastric banding arm proportional to increases in IS, with no net benefit on β-cell function. CONCLUSIONS Reducing demand on the β-cell by improving IS through weight loss does not reverse β-cell dysfunction. L + M was the only treatment that enhanced β-cell function.
Collapse
Affiliation(s)
- Kristina M. Utzschneider
- VA Puget Sound Health Care System, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - David A. Ehrmann
- Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Silva A. Arslanian
- UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Thomas A. Buchanan
- Department of Research & Evaluation, Kaiser Permanente Southern California, Pasadena, California, USA
| | - Sonia Caprio
- Department of Pediatrics, Yale University, New Haven, Connecticut, USA
| | | | - Tamara S. Hannon
- Department of Internal Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Steven E. Kahn
- VA Puget Sound Health Care System, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | | | - Kieren J. Mather
- Department of Internal Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Kristen J. Nadeau
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Susan Sam
- Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Mark Tripputi
- George Washington University Biostatistics Center, Rockville, Maryland, USA
| | - Anny H. Xiang
- Department of Research & Evaluation, Kaiser Permanente Southern California, Pasadena, California, USA
| | - Laure El ghormli
- George Washington University Biostatistics Center, Rockville, Maryland, USA
| | | |
Collapse
|
45
|
Tamborlane WV, Bishai R, Geller D, Shehadeh N, Al-Abdulrazzaq D, Vazquez EM, Karoly E, Troja T, Doehring O, Carter D, Monyak J, Sjöström CD. Once-Weekly Exenatide in Youth With Type 2 Diabetes. Diabetes Care 2022; 45:1833-1840. [PMID: 35679098 PMCID: PMC9346995 DOI: 10.2337/dc21-2275] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 04/01/2022] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Approved treatments for type 2 diabetes in pediatric patients include metformin, liraglutide, and insulin. However, approximately one-half of the youth fail metformin monotherapy within 1 year, insulin therapy is associated with challenges, and liraglutide requires daily injections. Consequently, the efficacy and safety of once-weekly injections of exenatide for the treatment of youth with type 2 diabetes was evaluated. RESEARCH DESIGN AND METHODS Participants (aged 10 to <18 years) were randomized (5:2) to once-weekly exenatide 2 mg or placebo, respectively. The primary efficacy end point was change in glycated hemoglobin from baseline to week 24. Secondary efficacy end points were also evaluated, and the frequency of adverse events (AEs) was assessed. RESULTS A total of 83 participants were randomized (exenatide, 59; placebo, 24) and 72 completed 24-week treatment (exenatide, 49; placebo, 23). At 24 weeks, the least squares mean change in glycated hemoglobin was -0.36% for the exenatide and +0.49% for the placebo groups (between-group difference, -0.85%; 95% CI -1.51, -0.19; P = 0.012). Nonsignificant least squares mean differences from baseline to 24 weeks favoring exenatide were observed: fasting glucose -21.6 mg/dL (-49.0, 5.7; P = 0.119), systolic blood pressure -2.8 mmHg (-8.0, 2.4; P = 0.284), and body weight -1.22 kg (-3.59, 1.15; P = 0.307). AEs occurred in 36 (61.0%) and 17 (73.9%) participants in the exenatide and placebo groups, respectively. CONCLUSIONS In youth with type 2 diabetes suboptimally controlled with current treatments, once-weekly exenatide reduced glycated hemoglobin at 24 weeks and was well tolerated.
Collapse
Affiliation(s)
| | - Raafat Bishai
- Late-Stage Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD
| | - David Geller
- The Center for Endocrinology, Diabetes, and Metabolism, Children’s Hospital Los Angeles, and Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Naim Shehadeh
- Bruce Rappaport Faculty of Medicine, Technion, and Institute of Diabetes and Endocrinology, Rambam Health Care Campus, Haifa, Israel
| | - Dalia Al-Abdulrazzaq
- Department of Population Health, Dasman Diabetes Institute, Dasman, Kuwait
- Department of Pediatrics, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | | | | | | | - Orlando Doehring
- Biometrics, Late-Stage Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, U.K
| | - Debra Carter
- Late-Stage Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD
| | - John Monyak
- Biometrics, Late-Stage Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD
| | - C. David Sjöström
- Late-Stage Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
46
|
Tricò D, Galderisi A, Van Name MA, Caprio S, Samuels S, Li Z, Galuppo BT, Savoye M, Mari A, Feldstein AE, Santoro N. A low n-6 to n-3 polyunsaturated fatty acid ratio diet improves hyperinsulinaemia by restoring insulin clearance in obese youth. Diabetes Obes Metab 2022; 24:1267-1276. [PMID: 35297549 PMCID: PMC9177628 DOI: 10.1111/dom.14695] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/05/2022] [Accepted: 03/16/2022] [Indexed: 11/29/2022]
Abstract
AIM To examine the determinants and metabolic impact of the reduction in fasting and postload insulin levels after a low n-6 to n-3 polyunsaturated fatty acid (PUFA) ratio diet in obese youth. MATERIALS AND METHODS Insulin secretion and clearance were assessed by measuring and modelling plasma insulin and C-peptide in 17 obese youth who underwent a nine-point, 180-minute oral glucose tolerance test (OGTT) before and after a 12-week, eucaloric low n-6:n-3 polyunsaturated fatty acid (PUFA) ratio diet. Hepatic fat content was assessed by repeated abdominal magnetic resonance imaging. RESULTS Insulin clearance at fasting and during the OGTT was significantly increased after the diet, while body weight, glucose levels, absolute and glucose-dependent insulin secretion, and model-derived variables of β-cell function were not affected. Dietary-induced changes in insulin clearance positively correlated with changes in whole-body insulin sensitivity and β-cell glucose sensitivity, but not with changes in hepatic fat. Subjects with greater increases in insulin clearance showed a worse metabolic profile at enrolment, characterized by impaired insulin clearance, β-cell glucose sensitivity, and glucose tolerance, and benefitted the most from the diet, achieving greater improvements in glucose-stimulated hyperinsulinaemia, insulin resistance, and β-cell function. CONCLUSIONS We showed that a 12-week low n-6:n-3 PUFA ratio diet improves hyperinsulinaemia by increasing fasting and postload insulin clearance in obese youth, independently of weight loss, glucose concentrations, and insulin secretion.
Collapse
Affiliation(s)
- Domenico Tricò
- Department of Surgical, Medical and Molecular Pathology and Critical Care MedicineUniversity of PisaPisa
| | | | - Michelle A. Van Name
- Department of PediatricsYale University School of MedicineNew HavenConnecticutUSA
| | - Sonia Caprio
- Department of PediatricsYale University School of MedicineNew HavenConnecticutUSA
| | - Stephanie Samuels
- Department of PediatricsYale University School of MedicineNew HavenConnecticutUSA
| | - Zhongyao Li
- Department of PediatricsYale University School of MedicineNew HavenConnecticutUSA
| | - Brittany T. Galuppo
- Department of PediatricsYale University School of MedicineNew HavenConnecticutUSA
| | - Mary Savoye
- Department of PediatricsYale University School of MedicineNew HavenConnecticutUSA
| | - Andrea Mari
- Institute of Neuroscience, National Research CouncilPaduaItaly
| | - Ariel E. Feldstein
- Department of PediatricsUniversity of California San DiegoSan DiegoCaliforniaUSA
| | - Nicola Santoro
- Department of PediatricsYale University School of MedicineNew HavenConnecticutUSA
- Department of Medicine and Health Sciences, “V.Tiberio” University of MoliseCampobassoItaly
| |
Collapse
|
47
|
Aroda VR, Krause-Steinrauf H, Kazemi EJ, Buse JB, Gulanski BI, Florez HJ, Ahmann AJ, Loveland A, Kuhn A, Lonier JY, Wexler DJ. Clinical and Metabolic Characterization of Adults With Type 2 Diabetes by Age in the Glycemia Reduction Approaches in Diabetes: A Comparative Effectiveness Study (GRADE) Cohort. Diabetes Care 2022; 45:1512-1521. [PMID: 40315175 PMCID: PMC9375440 DOI: 10.2337/dc21-2659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 04/09/2022] [Indexed: 05/04/2025]
Abstract
OBJECTIVE Differences in type 2 diabetes phenotype by age are described, but it is not known whether these differences are seen in a more uniformly defined adult population at a common early stage of care. We sought to characterize age-related clinical and metabolic characteristics of adults with type 2 diabetes on metformin monotherapy, prior to treatment intensification. RESEARCH DESIGN AND METHODS In the Glycemia Reduction Approaches in Diabetes: A Comparative Effectiveness Study (GRADE), participants were enrolled who had type 2 diabetes duration <10 years, had HbA1c 6.8-8.5%, and were on metformin monotherapy. Participants were randomly assigned to one of four additional glucose-lowering medications. We compared baseline clinical and metabolic characteristics across age categories (<45, 45 to <55, 55 to <65, and ≥65 years) using ANOVA and Pearson χ2 tests. RESULTS Within the GRADE cohort (n = 5,047), we observed significant differences by age, with younger adults having greater racial diversity, fewer medications for common comorbidities, lower prevalence of CVD, higher weight and BMI, and more pronounced hyperglycemia and diabetic dyslipidemia and with metabolic profile indicating lower insulin sensitivity (inverse fasting insulin [1/(fasting insulin)], HOMA of steady-state insulin sensitivity, Matsuda index) and inadequate β-cell response (oral disposition index) (P < 0.05 across age categories). CONCLUSIONS Clinical and metabolic characteristics of type 2 diabetes differ by age within the GRADE cohort. Younger adults exhibit more prominent obesity-related characteristics, including higher obesity levels and lower insulin sensitivity and β-cell compensation. Given the increasing burden of type 2 diabetes and complications, particularly among younger populations, these age-related distinctions may inform risk factor management approaches and treatment priorities. Further study will determine whether age-related differences impact response to therapy.
Collapse
Affiliation(s)
- Vanita R. Aroda
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- MedStar Health Research Institute, Hyattsville, MD
| | - Heidi Krause-Steinrauf
- The Biostatistics Center, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Rockville, MD
| | - Erin J. Kazemi
- The Biostatistics Center, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Rockville, MD
| | - John B. Buse
- University of North Carolina School of Medicine, Chapel Hill, NC
| | - Barbara I. Gulanski
- Endocrinology, Department of Medicine, Yale University School of Medicine, New Haven, CT
| | - Hermes J. Florez
- Department of Medicine, University of Miami, Miami, FL
- Geriatric Research Education and Clinical Center, Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, FL
- Medical University of South Carolina, Charleston, SC
| | - Andrew J. Ahmann
- Division of Endocrinology, Diabetes & Clinical Nutrition, Oregon Health & Science University, Portland, OR
| | - Amy Loveland
- MedStar Health Research Institute, Hyattsville, MD
| | | | - Jacqueline Y. Lonier
- Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY
| | - Deborah J. Wexler
- Diabetes Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
48
|
Galderisi A, Perilongo G, Caprio S, Da Dalt L, Di Salvo G, Gatta M, Giaquinto C, Rizzuto R, Robb A, Sly PD, Simonelli A, Staiano A, Vettor R, Baraldi E. Pediatric Preventive Care in Middle-High Resource Countries-The Padova Chart for Health in Children. Front Pediatr 2022; 10:803323. [PMID: 35498805 PMCID: PMC9047691 DOI: 10.3389/fped.2022.803323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 02/14/2022] [Indexed: 12/01/2022] Open
Abstract
Importance The Padova Chart for Health in Children (PCHC) aims to gather the evidence of healthcare promotion and protection for chidren and adolescents (i.e., aged <18 y) into a single document in order to guide families, healthcare providers and social actors on healthy choices. No more than 2% of Europeans and North Americans aged <30 y have a healthy lifestyle. This, together with metabolic and brain plasticity during childhood, creates the ideal opportunity to implement preventive strategies. Guided interventions promoting healthy lifestyle in children and families therefore have a key role in abating the unprecedented pandemic of non-communicable diseases (NCDs) in adulthood. Observations The PCHC is divided into four sections: nutrition, cardiovascular health, respiratory health, and mental and social health. Each section is structured in an ALICE approach (assessment, lobbying, intervention, call-for-action, evaluation): assessment of necessity, describing relevance to healthcare; lobbying to identify those who can effect the proposed interventions; interventions involving family, school and peers; a call-for-action to define priorities among the proposed interventions; and objective evaluation measures that can be applied on a population basis. Conclusions and Relevance Interventions promoting health in childhood require joint action from multiple institutional, local and family representatives, with the shared goal of promoting health across the entire age group. These lifestyle interventions have the potential to change the lifetime risk trajectory for NCDs.
Collapse
Affiliation(s)
- Alfonso Galderisi
- Department of Woman and Child's Health, University Hospital of Padova, Padova, Italy
| | - Giorgio Perilongo
- Department of Woman and Child's Health, University Hospital of Padova, Padova, Italy
| | - Sonia Caprio
- Department of Pediatrics, Yale University, New Haven, CT, United States
| | - Liviana Da Dalt
- Department of Woman and Child's Health, University Hospital of Padova, Padova, Italy
| | - Giovanni Di Salvo
- Department of Woman and Child's Health, University Hospital of Padova, Padova, Italy
| | - Michela Gatta
- Department of Woman and Child's Health, University Hospital of Padova, Padova, Italy
| | - Carlo Giaquinto
- Department of Woman and Child's Health, University Hospital of Padova, Padova, Italy
| | - Rosario Rizzuto
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Adelaide Robb
- Division of Psychiatry and Behavioral Sciences, Children's National Hospital, Washington, DC, United States
| | - Peter David Sly
- Children's Health and Environment Program, Child Health Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Alessandra Simonelli
- Department of Developmental Psychology and Socialization, University of Padova, Padova, Italy
| | - Annamaria Staiano
- Department of Translational Medical Science, Section of Pediatrics, University Federico II, Naples, Italy
| | - Roberto Vettor
- Department of Medicine, University Hospital of Padova, Padova, Italy
| | - Eugenio Baraldi
- Department of Woman and Child's Health, University Hospital of Padova, Padova, Italy
| |
Collapse
|
49
|
Shah AS, Nadeau KJ, Dabelea D, Redondo MJ. Spectrum of Phenotypes and Causes of Type 2 Diabetes in Children. Annu Rev Med 2022; 73:501-515. [PMID: 35084995 PMCID: PMC9022328 DOI: 10.1146/annurev-med-042120-012033] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Several factors, including genetics, family history, diet, physical activity, obesity, and insulin resistance in puberty, appear to increase the risk of type 2 diabetes in youth. Youth-onset type 2 diabetes is often thought of as a single entity but rather exists as a spectrum of disease with differences in presentation, metabolic characteristics, clinical progression, and complication rates. We review what is currently known regarding the risks associated with developing type 2 diabetes in youth. Additionally, we focus on the spectrum of phenotypes of pediatric type 2 diabetes, discuss the pathogenic underpinnings and potential therapeutic relevance of this heterogeneity, and compare youth-onset type 2 diabetes with type 1 diabetes and adult-onset type 2 diabetes. Finally, we highlight knowledge gaps in prediction and prevention of youth-onset type 2 diabetes.
Collapse
Affiliation(s)
- Amy S. Shah
- Cincinnati Children’s Hospital Medical Center and University of Cincinnati, Cincinnati, Ohio 45229, USA
| | - Kristen J. Nadeau
- Children’s Hospital Colorado and University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA
| | - Dana Dabelea
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Department of Epidemiology, and Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA
| | - Maria J. Redondo
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas 77030, USA
| |
Collapse
|
50
|
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
|